1
|
Bjerre-Bastos JJ, Sejersen C, Nielsen HB, Boesen M, Secher NH, Distajo G, Flood V, Henrotin Y, Uebelhoer M, Krustrup P, Kitchen CC, Thudium CS, Andersen JR, Bihlet AR. The Impact of Weight-bearing Exercise, Non-Weight-bearing Exercise, and Cardiovascular Stress on Biochemical Markers of Cartilage Turnover in Patients With Mild to Moderate Knee Osteoarthritis - A Sequential, Cross-Over, Clinical Study. Cartilage 2025; 16:159-168. [PMID: 38853398 PMCID: PMC11569697 DOI: 10.1177/19476035241258170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/11/2024] Open
Abstract
ObjectiveTo investigate how running, cycling, and sedentary cardiovascular stress impact biomarkers of cartilage turnover acutely in subjects with knee osteoarthritis (OA).DesignThis was a sequential, cross-over, clinical study. Forty subjects with primary knee OA underwent moderate-to-high-intensity cycling, running, and adrenaline infusion on separate days. Blood was sampled before, during, and at 6-time points after intervention. On a control day, similar samples were taken. Biomarkers of type II collagen degradation (C2M, T2CM, Coll2-1, Coll2-1NO2), formation (PRO-C2), and aggrecan degradation (ARGS) were measured.ResultsMean age was 60.4 years, 40% were male, 45% had cumulated Kellgren-Lawrence (KL)-grade (Right + Left knee) of 2 to 3 and 55% had 4 to 6. Analyzing overall changes, area under the curve was significantly lower compared with resting values for ARGS and C2M after cycling and for ARGS after running. Considering individual time points, peak changes in biomarker levels showed reduction in C2M shortly following cycling (T20min = -12.3%, 95% confidence interval [CI]: -19.3% to -5.2%). PRO-C2 increased during cycling (T10min = 14.0%, 95% CI = 4.1% to 23.8%) and running (T20min = 16.5%, 95% CI = 4.3% to 28.6%). T2CM decreased after cycling (T50min = -19.9%, 95% CI = -29.2% to -10.6%), running (T50min = -22.8%, 95% CI = -32.1% to -13.5%), and infusion of adrenaline (peak, T50min = -9.8%, 95% CI = -20.0% to 0.4%). A latent increase was seen in Coll2-1 240 minutes after running (T260min = 21.7%, 95% CI = -1.6% to 45.1%).ConclusionExercise had an impact on cartilage markers, but it did not suggest any detrimental effect on cartilage. Changes following adrenaline infusion suggest a sympathomimetic influence on the serological composition of biomarkers.
Collapse
Affiliation(s)
- Jonathan J. Bjerre-Bastos
- Xlab, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- NBCD A/S, Herlev, Denmark
| | - Casper Sejersen
- Department of Anaesthesiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henning Bay Nielsen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Anaesthesia and Intensive Care Medicine, Zealand University Hospital, Roskilde, Denmark
- Department of Nutrition, Exercise and Sport, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Boesen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Radiology, Bispebjerg and Frederiksberg Hospitals, Copenhagen, Denmark
| | - Niels H. Secher
- Department of Anaesthesiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Yves Henrotin
- Department of Physical and Rehabilitation, VIVALIA, Marche-en-Famenne, Belgium
- Musculoskeletal Innovative Research Lab (mSKIL), University of Liège, Liège, Belgium
| | | | - Peter Krustrup
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Carl-Christian Kitchen
- Department of Anaesthesiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
2
|
Clark SJ, Curcio C, Dick AD, Doyle S, Edwards M, Flores-Bellver M, Hass D, Lennon R, Toomey CB, Rohrer B. Breaking Bruch's: How changes in Bruch's membrane influence retinal homeostasis. Exp Eye Res 2025; 255:110343. [PMID: 40107443 DOI: 10.1016/j.exer.2025.110343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Affiliation(s)
- Simon J Clark
- Institute for Ophthalmic Research, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Christine Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham Heersink School of Medicine, USA
| | - Andrew D Dick
- University of Bristol and UCL-Institute of Ophthalmology and NIHR Biomedical Research Centre, Moorfields Eye Hospital and UCL-Institute of Ophthalmology, UK
| | - Sarah Doyle
- Department of Clinical Medicine, School of Medicine and Trinity Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Malia Edwards
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Miguel Flores-Bellver
- Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Daniel Hass
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Christopher B Toomey
- Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California at San Diego, La Jolla, CA, USA
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston SC, USA.
| |
Collapse
|
3
|
Farrell M, Waibel FWA, Carrera I, Spattini G, Clark L, Adams RJ, Von Pfeil DJF, De Sousa RJR, Villagrà DB, Amengual-Vila M, Paviotti A, Quinn R, Harper J, Clarke SP, Jordan CJ, Hamilton M, Moores AP, Greene MI. Musculoskeletal adverse events in dogs receiving bedinvetmab (Librela). Front Vet Sci 2025; 12:1581490. [PMID: 40417367 PMCID: PMC12100767 DOI: 10.3389/fvets.2025.1581490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/04/2025] [Indexed: 05/27/2025] Open
Abstract
Objectives To conduct a specialist-led disproportionality analysis of musculoskeletal adverse event reports (MSAERs) in dogs treated with bedinvetmab (Librela™) compared to six comparator drugs with the same indication. Furthermore, to report the findings from a subset of dogs whose adverse event (AE) data underwent independent adjudication by an expert panel. Study design Case-control study and case series analysis. Sample population The European Medicines Agency's EudraVigilance database (2004-2024) and 19 client-owned dogs. Methods An EBVS® Veterinary Specialist in Surgery individually reviewed all MSAERs to Librela™, Rimadyl®, Metacam®, Previcox®, Onsior®, Galliprant®, and Daxocox® (2004-2024). The primary null hypothesis was that Librela's MSAER rate would not exceed that of comparator drugs by more than 50%. The secondary hypothesis was that MSAER would surge and taper following the launch of new drugs. Results The disproportionality analysis did not support the hypotheses. Ligament/tendon injury, polyarthritis, fracture, musculoskeletal neoplasia, and septic arthritis were reported ~9-times more frequently in Librela-treated dogs than the combined total of dogs treated with the comparator drugs. A review of 19 suspected musculoskeletal adverse events (MSAEs) by an 18-member expert panel unanimously concluded a strong suspicion of a causal association between bedinvetmab and accelerated joint destruction. Conclusion This study supports recent FDA analyses by demonstrating an increased reporting rate of musculoskeletal adverse events in dogs treated with Librela. Further investigation and close clinical monitoring of treated dogs are warranted. Impact Our findings should serve as a catalyst for large-scale investigations into bedinvetmab's risks and pharmacovigilance.
Collapse
Affiliation(s)
- Mike Farrell
- Ferguson Veterinary Clinic, Glenrothes, United Kingdom
| | - Felix W. A. Waibel
- Department of Orthopaedic Surgery, Balgrist University Hospital, Zurich, Switzerland
| | | | | | - Louise Clark
- Davies Veterinary Specialists, Shillington, United Kingdom
| | | | | | | | | | | | | | - Rob Quinn
- Vita Referrals, Wetherby, United Kingdom
| | - Justin Harper
- Texas Specialty Veterinary Services, Boerne, TX, United States
| | | | | | | | - Andy P. Moores
- The Moores Orthopaedic Clinic, Basingstoke, United Kingdom
| | - Mark Irwin Greene
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
4
|
Pueyo Moliner A, Ito K, Zaucke F, Kelly DJ, de Ruijter M, Malda J. Restoring articular cartilage: insights from structure, composition and development. Nat Rev Rheumatol 2025; 21:291-308. [PMID: 40155694 DOI: 10.1038/s41584-025-01236-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 04/01/2025]
Abstract
Articular cartilage can withstand substantial compressive and shear forces within the joint and also reduces friction during motion. The exceptional mechanical properties of articular cartilage stem from its highly organized extracellular matrix (ECM). The ECM is composed mainly of collagen type II and is pivotal in conferring mechanical durability to the tissue within its proteoglycan-rich matrix. Articular cartilage is prone to injury and degeneration, and current treatments often fail to restore the mechanical function of this tissue. A key challenge is replicating the intricate collagen-proteoglycan network, which is essential for the long-lasting restoration and mechanical durability of the tissue. Understanding articular cartilage development, which arises between late embryonic and early juvenile development, is vital for the creation of durable therapeutic strategies. The development of the articular ECM involves the biosynthesis, fibrillogenesis and self-assembly of the collagen type II network, which, along with proteoglycans and minor ECM components, shapes the architecture of adult articular cartilage. A deeper understanding of these processes could inform biomaterial-based therapies aimed at improving therapeutic outcomes. Emerging biofabrication technologies offer new opportunities to integrate developmental principles into the creation of durable articular cartilage implants. Bridging fundamental biology with innovative engineering offers novel approaches to generating more-durable 3D implants for articular cartilage restoration.
Collapse
Affiliation(s)
- Alba Pueyo Moliner
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Keita Ito
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Frank Zaucke
- Department of Trauma Surgery and Orthopedics, Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mylène de Ruijter
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Jos Malda
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands.
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands.
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
5
|
Hung CY, Hsueh TY, Rethi L, Lu HT, Chuang AEY. Advancements in regenerative medicine: a comprehensive review of stem cell and growth factor therapies for osteoarthritis. J Mater Chem B 2025; 13:4494-4526. [PMID: 40042377 DOI: 10.1039/d4tb01769b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Osteoarthritis (OA) is a widely encountered degenerative joint disorder marked by gradual cartilage deterioration, inflammation, and pain, which collectively impose considerable strain on global healthcare systems. While traditional therapies typically offer relief from symptoms, they do not tackle the core pathophysiological aspects of the disease. Regenerative medicine has recently risen as a promising field for addressing OA, capitalizing on the regenerative capabilities of stem cells and growth factors to foster tissue healing and renewal. This thorough review delves into the most recent progress in stem cell and growth factor treatments for OA, covering preclinical studies, clinical trials, and novel technological developments. We discuss the diverse origins of stem cells, such as mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and adipose-derived stem cells (ASCs), underscoring their therapeutic actions and effectiveness in both preclinical and clinical environments. Moreover, we explore contributions of growth factors like transforming growth factor (TGF)-β, platelet-derived growth factor (PDGF), and insulin-like growth factor (IGF) in modifying OA's pathology and enhancing tissue restoration. Additionally, this review discusses the hurdles and constraints tied to current regenerative strategies, including the standardization of cell sources, the refinement of delivery techniques, and considerations for long-term safety. By meticulously assessing the latest research outcomes and technological breakthroughs, this review aims to shed light on the potential of stem cell and growth factor therapies as forthcoming therapeutic options for OA, thereby propelling forward the domain of regenerative medicine and enhancing clinical results for individuals afflicted with this incapacitating ailment.
Collapse
Affiliation(s)
- Chen-Yuan Hung
- School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tai-Yuan Hsueh
- School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Lekshmi Rethi
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan
| | - Hsien-Tsung Lu
- Department of Orthopedics, Taipei Medical University Hospital, Taipei City 11031, Taiwan
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- International PhD Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Andrew E-Y Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan
- Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| |
Collapse
|
6
|
Anderson-Watters M, Khan IM. BMP9 induces postnatal zonal stratification of immature articular cartilage through reconfiguration of the existing collagen framework. Front Cell Dev Biol 2025; 12:1511908. [PMID: 39935787 PMCID: PMC11810917 DOI: 10.3389/fcell.2024.1511908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/04/2024] [Indexed: 02/13/2025] Open
Abstract
Articular cartilage lines bones in synovial joints, and its main structural element, collagen, has an arcade-like arrangement formed from an initially random network in a process called postnatal maturation. This reshaping of the extracellular matrix is similar across all species and is critical for the lifelong strength and durability of cartilage. Collagen remodelling during maturation is difficult to study because it spans a period of time between birth and puberty, and in larger animals this can be months or years. In this study, we show that growth factor bone morphogenetic protein-9 (BMP9) induces collagen remodelling in intact immature articular cartilage explants within 21 days, generating the characteristic arcade-like structure and zonal anisotropic architecture of adult cartilage. In explants exposed to BMP9, collagen fibrils underwent angular displacement from 19° to 78° with respect to the surface, cell density decreased 1.77-fold, and chondrons were significantly larger. The absence of labelling with anti-COL2¾m, a marker of collagen turnover, showed that the existing fibril network was restructured. We found that stromelysin-1 (metalloproteinase-3, MMP3) gene expression was consistently upregulated, whilst other MMP transcript levels were unchanged or reduced. Remodelling was dependent on proteoglycan turnover and could be inhibited using PD166973. These data suggest a possible mechanism whereby MMP3 induces proteoglycan turnover and depolymerises collagen fibrils enabling them to undergo spatial reorganisation. This process may be driven by tissue swelling, which generates directional strain to align fibrils into an arcade-like pattern. The ability to induce tissue maturation advances the potential for engineering durable and functional cartilage for patients requiring joint repair due to diseases such as osteoarthritis.
Collapse
Affiliation(s)
| | - Ilyas M. Khan
- Faculty of Medicine, Health and Life Science, Swansea University, Swansea, United Kingdom
| |
Collapse
|
7
|
Tsuchiya Y, Yeung CYC, Svensson RB, Kjaer M. Effect of human myoblasts on tenogenic progression in 2D and 3D culture models. J Anat 2025. [PMID: 39854094 DOI: 10.1111/joa.14224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/04/2024] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
Tendon injuries and disorders associated with mechanical tendon overuse are common musculoskeletal problems. Even though tendons play a central role in human movement, the intrinsic healing process of tendon is very slow. So far, it is known that tendon cell activity is supported by several interstitial cells within the tendon. However, the interplay between the tendon and the adjacent muscle for tendon regeneration and development processes has not been fully investigated. Here, we tested whether factors released from muscle derived myogenic cells (myoblasts) enhance tenogenic progressions of human tendon derived cells (tendon fibroblasts) using two-dimensional (2D) culture model and a three-dimensional (3D)-engineered tendon construct culture model, which mimics tendon regeneration and development. The conditioned media from myoblasts and unconditioned media as control were applied to tendon fibroblasts. In 2D, immunofluorescence analysis revealed increased collagen type I expressing area and increased migration potential when conditioned media from myoblasts were applied. In the 3D-engineered human tendon construct model, wet weight, diameter, and cross-sectional area of the tendon constructs were increased in response to the application of conditioned media from myoblasts, whereas the collagen density was lower and mechanical function was reduced both at the functional level (maximum stiffness) and the material level (maximum stress and modulus). These results indicate that myoblast-derived factors extend collagen expressing area and enhance migration of tendon fibroblasts, while factors involved in the robustness of extra-cellular matrix deposition of tissue-engineered tendon constructs are lacking. Our findings suggest that adjacent muscle affects the signaling interplay in tendons.
Collapse
Affiliation(s)
- Yoshifumi Tsuchiya
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg-Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Sports Science, Doshisha University, Kyoto, Japan
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Japan
| | - Ching-Yan Chloé Yeung
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg-Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Rene B Svensson
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg-Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg-Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Kauppinen S, Fercher D, Barreto G, Karjalainen VP, Virtanen V, Baixauli-Marin L, Fonti M, Zhang S, Frondelius T, Weber P, Saarakkala S, Zenobi-Wong M, Finnilä MAJ. Assessment of whole cartilage surface damage in an osteoarthritis rat model: The Cartilage Roughness Score (CRS) utilizing microcomputed tomography. Osteoarthritis Cartilage 2025; 33:134-145. [PMID: 39357597 DOI: 10.1016/j.joca.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/27/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE This study aims to establish an accurate and robust imaging biomarker for pre-clinical osteoarthritis (OA) research, focusing on early detection of cartilage surface degeneration. METHOD Using 50 male Wistar rats, this study aims to observe Collagenase-induced OA (CIOA) progression through microcomputed x-ray tomography (µCT), histopathological analysis, and gait analysis. A novel parameter, Cartilage Roughness Score (CRS), was developed for assessing cartilage structural damage from µCT data and was compared with histological OARSI Cartilage Degeneration Score (OARSI CDS). Additionally, as CRS maps the full surface, it was used to simulate the level of uncertainty in histological sampling. RESULTS CRS and OARSI CDS have a linear relationship. CRS for healthy cartilage is 2.75 (95% CI: 1.14-4.36), and with every 1 unit increase in OARSI, CRS is expected to increase by 0.64 (95% CI: 0.35-0.92). Cartilage degeneration due to CIOA was evident in both histopathological scoring and CRS. However, only CRS was sensitive enough to show consistent damage progression from day 10 to day 60. Furthermore, our simulation for histological sampling suggested that up to 16 coronal slices with 200 µm spacing would be needed to accurately represent the full extent of cartilage surface degeneration in a slice-wise manner. Gait analysis showed changes solely at eight days post-collagenase injection, normalizing by day 60. CONCLUSION The CRS analysis method emerges as a robust tool for cartilage surface damage assessment. This study demonstrates the potential of automatic 3D analysis over the traditional 2D histological approach when evaluating cartilage surface damage.
Collapse
Affiliation(s)
- Sami Kauppinen
- Research unit of Health Sciences and Technology, University of Oulu, Finland.
| | - David Fercher
- Tissue Engineering and Biofabrication, ETH Zürich, Switzerland
| | - Gonçalo Barreto
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Vesa Virtanen
- Research unit of Health Sciences and Technology, University of Oulu, Finland
| | | | - Marina Fonti
- Tissue Engineering and Biofabrication, ETH Zürich, Switzerland
| | - Shipin Zhang
- Tissue Engineering and Biofabrication, ETH Zürich, Switzerland
| | - Tuomas Frondelius
- Research unit of Health Sciences and Technology, University of Oulu, Finland
| | - Patrick Weber
- Tissue Engineering and Biofabrication, ETH Zürich, Switzerland
| | - Simo Saarakkala
- Research unit of Health Sciences and Technology, University of Oulu, Finland
| | | | - Mikko A J Finnilä
- Research unit of Health Sciences and Technology, University of Oulu, Finland
| |
Collapse
|
9
|
Fugazzola MC, De Ruijter M, Veraa S, Plomp S, van Buul W, Hermsen G, van Weeren R. A hybrid repair strategy for full-thickness cartilage defects: Long-term experimental study in eight horses. J Orthop Res 2025; 43:59-69. [PMID: 39292194 PMCID: PMC11615427 DOI: 10.1002/jor.25972] [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: 04/03/2024] [Revised: 07/19/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
The objective of this study was to evaluate a non-resorbable implant for the focal repair of chondral defects in eight adult horses with 12-month follow-up. The bi-layered construct composed of a polycarbonate-urethane-urea biomaterial which was printed in 3D fashion onto a bone anchor was implanted into surgically created osteochondral defects into the femoropatellar joints of eight horses. The analysis of post-mortem outcomes were compared to defects treated with microfracture in the same animal on the contralateral femoropatellar jointfemoropatellar joint. The overall macroscopic scoring after 12 months yielded higher scores in the OCI-treated stifles compared to MF treatment (p = 0.09) with better quality and filling of the defect. Histology revealed good anchorage of repair tissue growing into the 3D structure of the implant and histopathology scoring for adjacent native cartilage showed no difference between groups. MRI and micro-CT showed overall less sclerotic reactions in the surrounding bone in the implant group and no foreign body reaction was detected. Biomechanical analysis of the repair tissue revealed a significantly higher peak modulus (p < 0.05) in the implant group (0.74 ± 0.45) compared to the microfracture control group (0.15 ± 0.11). Dynamic loading yielded higher values for the repair tissue overgrowing the implant group (0.23 ± 0.17) compared to the microfracture control (0.06 ± 0.06) (p < 0.05). The bi-layered osteochondral implant provided a safe implant for focal repair of full-thickness osteochondral defects, as no adverse reaction was seen within the joints and the level of degeneration of adjacent cartilage to the repair site was not different compared to that seen in defects treated with microfracture after 12 months.
Collapse
Affiliation(s)
- Maria C. Fugazzola
- Department of Clinical Sciences, Faculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
| | - Mylène De Ruijter
- Department of Clinical Sciences, Faculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
- Department of OrthopedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Stefanie Veraa
- Department of Clinical Sciences, Faculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
| | - Saskia Plomp
- Department of Clinical Sciences, Faculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
- Department of OrthopedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | | | - René van Weeren
- Department of Clinical Sciences, Faculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
10
|
Latourte A, Jaulerry S, Combier A, Cherifi C, Jouan Y, Grange T, Daligault J, Ea HK, Cohen-Solal M, Hay E, Richette P. SerpinA3N limits cartilage destruction in osteoarthritis by inhibiting macrophage-derived leucocyte elastase. Ann Rheum Dis 2024; 83:1781-1790. [PMID: 39134394 DOI: 10.1136/ard-2024-225645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/31/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Inflammatory mediators such as interleukin 6 (IL-6) are known to activate catabolic responses in chondrocytes during osteoarthritis (OA). This study aimed to investigate the role of a downstream target gene of IL-6, the serine protease inhibitor SerpinA3N, in the development of cartilage damage in OA. METHODS RNA sequencing was performed in murine primary chondrocytes treated with IL-6, and identified target genes were confirmed in human and murine OA cartilage samples. Male cartilage-specific Serpina3n-deficient mice and control mice underwent meniscectomy (MNX) or sham surgery at 10 weeks of age. Intra-articular injections of SerpinA3N or sivelestat (an inhibitor of leucocyte elastase (LE), a substrate for SerpinA3N) were performed in wild-type mice after MNX. Joint damage was assessed 3-9 weeks after surgery by histology and micro-CT. The effect of sivelestat was assessed in cartilage explants exposed to macrophage-derived conditioned media. RESULTS RNA sequencing revealed that SerpinA3N is a major target gene of IL-6 in chondrocytes. The expression of SerpinA3N is increased in OA cartilage. Conditional loss of SerpinA3N in chondrocytes aggravated OA in mice, while intra-articular injection of SerpinA3N limited joint damage. Chondrocytes did not produce serine proteases targeted by SerpinA3N. By contrast, macrophages produced LE on IL-6 stimulation. Sivelestat limited the cartilage catabolism induced by conditioned media derived from IL-6-stimulated macrophages. Additionally, an intra-articular injection of sivelestat is protected against OA in the MNX model. CONCLUSIONS SerpinA3N protects cartilage against catabolic factors produced by macrophages, including LE. SerpinA3N and LE represent new therapeutic targets to dampen cartilage damage in OA.
Collapse
Affiliation(s)
- Augustin Latourte
- Inserm UMR-S 1132, Université Paris Cité, Paris, France
- Rheumatology Department, Hôpital Lariboisière APHP.Nord, Paris, France
| | - Sarah Jaulerry
- Inserm UMR-S 1132, Université Paris Cité, Paris, France
- Rheumatology Department, Hôpital Lariboisière APHP.Nord, Paris, France
| | - Alice Combier
- Inserm UMR-S 1132, Université Paris Cité, Paris, France
| | | | - Yohan Jouan
- Inserm UMR-S 1132, Université Paris Cité, Paris, France
| | - Thierry Grange
- Institut Jacques Monod, Université Paris Cité, Paris, Île-de-France, France
| | - Julien Daligault
- Institut Jacques Monod, Université Paris Cité, Paris, Île-de-France, France
| | - Hang-Korng Ea
- Inserm UMR-S 1132, Université Paris Cité, Paris, France
- Rheumatology Department, Hôpital Lariboisière APHP.Nord, Paris, France
| | - Martine Cohen-Solal
- Inserm UMR-S 1132, Université Paris Cité, Paris, France
- Rheumatology Department, Hôpital Lariboisière APHP.Nord, Paris, France
| | - Eric Hay
- Inserm UMR-S 1132, Université Paris Cité, Paris, France
| | - Pascal Richette
- Inserm UMR-S 1132, Université Paris Cité, Paris, France
- Rheumatology Department, Hôpital Lariboisière APHP.Nord, Paris, France
| |
Collapse
|
11
|
Callan KT, Otarola G, Brown WE, Athanasiou KA, Wang D. The Longer-Term Effects of a Single Bupivacaine Exposure on the Mechanical Properties of Native Cartilage Explants. Cartilage 2024; 15:156-163. [PMID: 36992533 PMCID: PMC11368901 DOI: 10.1177/19476035231164751] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/04/2023] [Accepted: 03/04/2023] [Indexed: 03/31/2023] Open
Abstract
OBJECTIVE The purpose of this study was to determine the in vitro effects of a single exposure of bupivacaine on the mechanical properties of bovine cartilage explants at 3 weeks. DESIGN Femoral condyle articular cartilage explants were aseptically harvested from juvenile bovine stifle joints before being exposed to chondrogenic medium containing 0.50% (wt/vol) bupivacaine, 0.25% (wt/vol) bupivacaine, or no medication (control) for 1 hour. Explants were then washed and maintained in culture in vitro for 3 weeks before testing. Cell viability, tensile and compressive mechanical properties, histological properties, and biochemical properties were then assessed. RESULTS Explants exhibited a dose-dependent decrease in mean tensile Young's modulus with increasing bupivacaine concentration (9.86 MPa in the controls, 6.48 MPa in the 0.25% bupivacaine group [P = 0.048], and 4.72 MPa in the 0.50% bupivacaine group [P = 0.005]). Consistent with these results, collagen content and collagen crosslinking decreased with bupivacaine exposure as measured by mass spectrometry. Compressive properties of the explants were unaffected by bupivacaine exposure. Explants also exhibited a trend toward dose-dependent decreases in viability (51.2% for the controls, 47.3% for the 0.25% bupivacaine-exposed group, and 37.0% for the 0.50% bupivacaine-exposed group [P = 0.072]). CONCLUSIONS Three weeks after 1-hour bupivacaine exposure, the tensile properties of bovine cartilage explants were significantly decreased, while the compressive properties remained unaffected. These decreases in tensile properties corresponded with reductions in collagen content and crosslinking of collagen fibers. Physicians should be judicious regarding the intra-articular administration of bupivacaine in native joints.
Collapse
Affiliation(s)
- Kylie T. Callan
- University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Gaston Otarola
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Wendy E. Brown
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | | | - Dean Wang
- University of California, Irvine School of Medicine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Department of Orthopaedic Surgery, University of California Irvine Health, Orange, CA, USA
| |
Collapse
|
12
|
Ignatyeva N, Gavrilov N, Timashev PS, Medvedeva EV. Prg4-Expressing Chondroprogenitor Cells in the Superficial Zone of Articular Cartilage. Int J Mol Sci 2024; 25:5605. [PMID: 38891793 PMCID: PMC11171992 DOI: 10.3390/ijms25115605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/12/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Joint-resident chondrogenic precursor cells have become a significant therapeutic option due to the lack of regenerative capacity in articular cartilage. Progenitor cells are located in the superficial zone of the articular cartilage, producing lubricin/Prg4 to decrease friction of cartilage surfaces during joint movement. Prg4-positive progenitors are crucial in maintaining the joint's structure and functionality. The disappearance of progenitor cells leads to changes in articular hyaline cartilage over time, subchondral bone abnormalities, and the formation of ectopic ossification. Genetic labeling cell technology has been the main tool used to characterize Prg4-expressing progenitor cells of articular cartilage in vivo through drug injection at different time points. This technology allows for the determination of the origin of progenitor cells and the tracking of their progeny during joint development and cartilage damage. We endeavored to highlight the currently known information about the Prg4-producing cell population in the joint to underline the significance of the role of these cells in the development of articular cartilage and its homeostasis. This review focuses on superficial progenitors in the joint, how they contribute to postnatal articular cartilage formation, their capacity for regeneration, and the consequences of Prg4 deficiency in these cells. We have accumulated information about the Prg4+ cell population of articular cartilage obtained through various elegantly designed experiments using transgenic technologies to identify potential opportunities for further research.
Collapse
Affiliation(s)
- Nadezda Ignatyeva
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119048, Russia; (N.G.); (P.S.T.); (E.V.M.)
| | | | | | | |
Collapse
|
13
|
Lee SJ, Jeon O, Lee YB, Alt DS, Ding A, Tang R, Alsberg E. In situ cell condensation-based cartilage tissue engineering via immediately implantable high-density stem cell core and rapidly degradable shell microgels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.20.590385. [PMID: 38712035 PMCID: PMC11071421 DOI: 10.1101/2024.04.20.590385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Formation of chondromimetic human mesenchymal stem cells (hMSCs) condensations typically required in vitro culture in defined environments. In addition, extended in vitro culture in differentiation media over several weeks is usually necessary prior to implantation, which is costly, time consuming and delays clinical treatment. Here, this study reports on immediately implantable core/shell microgels with a high-density hMSC-laden core and rapidly degradable hydrogel shell. The hMSCs in the core formed cell condensates within 12 hours and the oxidized and methacrylated alginate (OMA) hydrogel shells were completely degraded within 3 days, enabling spontaneous and precipitous fusion of adjacent condensed aggregates. By delivering transforming growth factor-β1 (TGF-β1) within the core, the fused condensates were chondrogenically differentiated and formed cartilage microtissues. Importantly, these hMSC-laden core/shell microgels, fabricated without any in vitro culture, were subcutaneously implanted into mice and shown to form cartilage tissue via cellular condensations in the core after 3 weeks. This innovative approach to form cell condensations in situ without in vitro culture that can fuse together with each other and with host tissue and be matured into new tissue with incorporated bioactive signals, allows for immediate implantation and may be a platform strategy for cartilage regeneration and other tissue engineering applications.
Collapse
Affiliation(s)
- Sang Jin Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, 909 S. Wolcott Ave., Chicago, IL, 60612 USA
| | - Oju Jeon
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, 909 S. Wolcott Ave., Chicago, IL, 60612 USA
| | - Yu Bin Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, 909 S. Wolcott Ave., Chicago, IL, 60612 USA
| | - Daniel S. Alt
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106 USA
| | - Aixiang Ding
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, 909 S. Wolcott Ave., Chicago, IL, 60612 USA
| | - Rui Tang
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, 909 S. Wolcott Ave., Chicago, IL, 60612 USA
| | - Eben Alsberg
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL 60612, USA
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, 909 S. Wolcott Ave., Chicago, IL, 60612 USA
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106 USA
- Departments of Mechanical & Industrial Engineering, Orthopaedic Surgery, and Pharmacology and Regenerative Medicine, University of Illinois at Chicago, 909 S. Wolcott Ave., Chicago, IL, 60612 USA
| |
Collapse
|
14
|
Bjerre-Bastos JJ, Hamrouni N, Henrotin Y, Thudium CS, Bihlet AR. Joint biomarker response to mechanical stimuli in osteoarthritis - A scoping review. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100390. [PMID: 37885822 PMCID: PMC10598037 DOI: 10.1016/j.ocarto.2023.100390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 08/04/2023] [Indexed: 10/28/2023] Open
Abstract
Objective Arthritic cartilage is primed for mechanical damage. Joint biochemical markers (JBM) could provide insight into the impact of mechanical stimulation on joint tissue turnover in osteoarthritis (OA) of potential use in clinical OA research and practice. However, existing studies of the acute impact of physical activities (PA) on JBM often contain risks of substantial bias. The purpose of this scoping review was to critically review and discuss existing reports of acute joint tissue turnover as reflected in JBM in relation to PA in OA and propose considerations for future research. Design We searched PubMed, Embase, and Scopus and reference lists for original reports on the acute impact of PA on JBM in human OA. Identified studies were reviewed by two reviewers forming the basis for the discussion of methodology. Results Search in databases resulted in nine eligible papers after full-text evaluation. Two additional papers were identified through reference lists, resulting in 11 papers included in this review. Ten investigated knee OA and one investigated hand OA. Biomarkers described were related to turnover of type II collagen, aggrecan, and cartilage oligomeric matrix protein. Conclusions The literature is dominated by small, simplistic studies, but suggests that mechanical stimulation can induce acute changes in joint biomarkers. In order to diminish the existing bias in future studies, it is important to recognize methodological considerations e.g. patient and biomarker selection as well as peri-interventional control. Common potential sources of bias include the acute shift in plasma volume due to cardiovascular stress and postural changes.
Collapse
Affiliation(s)
- Jonathan J. Bjerre-Bastos
- Xlab, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- NBCD A/S, Herlev, Denmark
- Department of Orthopedic Surgery, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Nizar Hamrouni
- Department of Orthopedic Surgery, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Yves Henrotin
- Musculoskeletal Innovative Research Lab (mSKIL), Arthropôle Liège, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
- Physical Therapy and Rehabilitation Department, Princess Paola Hospital, Vivalia, Rue du Vivier, 6900 Marche-en-Famenne, Belgium
- Artialis SA, CHU Sart-Tilman, GIGA Tower, level 3, 4000 Liège, Belgium
| | | | | |
Collapse
|
15
|
Bai X, Sun H, Jia L, Xu J, Zhang P, Zhang D, Gu Y, Chen B, Feng L. Chondrocyte targeting gold nanoparticles protect growth plate against inflammatory damage by maintaining cartilage balance. Mater Today Bio 2023; 23:100795. [PMID: 37766899 PMCID: PMC10519832 DOI: 10.1016/j.mtbio.2023.100795] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/09/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Cartilage destruction caused by inflammation is a clinical challenge. Many studies have investigated cartilage destruction in adults, but little research was conducted on children. In this study, the protective effect of gold nanoparticles (AuNPs) on the cartilage of children was realized by counteracting chondrocyte apoptosis and extracellular matrix (ECM) degradation in a young mouse model of lipopolysaccharide (LPS)-induced growth plate (GP) cartilage damage. Initially, engineered AuNPs can be efficiently absorbed by chondrocytes, approximately 20 times the amount absorbed by macrophages, resulting in a 29% ± 0.05% increase in chondrocyte viability. Then, AuNPs exposure significantly reduced the release of inflammatory cytokines and secretion of ECM degradation factors induced by LPS. Subsequently, AuNPs were applied to resist LPS-induced cartilage destruction in young mice. AuNPs inhibited the formation of gaps, without chondrocytes and extracellular matrix, between the proliferative and hypertrophy zones of the GP cartilage, and the gaps were noticeable in the LPS group. This finding can be attributed to the capability of AuNPs to reduce the LPS-induced apoptosis rate of mouse chondrocytes by 72.38% and the LPS-induced ECM degradation rate by 70.89%. Further analysis demonstrated that remission is partly due to AuNPs' role in maintaining the balance of catabolic and anabolic factors in the ECM. Altogether, these findings indicate that AuNPs can partially protect the cartilage of children from inflammatory damage by suppressing chondrocyte apoptosis and ECM degradation.
Collapse
Affiliation(s)
- Xue Bai
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Basic Research in Clinical Applied Biomechanics, China
| | - Hongyan Sun
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Lina Jia
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Junjie Xu
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Peng Zhang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Deyuan Zhang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100083, China
| | - Yu Gu
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Basic Research in Clinical Applied Biomechanics, China
| | - Bo Chen
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Lin Feng
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100083, China
| |
Collapse
|
16
|
Ruscitto A, Chen P, Tosa I, Wang Z, Zhou G, Safina I, Wei R, Morel MM, Koch A, Forman M, Reeve G, Lecholop MK, Wilson M, Bonthius D, Chen M, Ono M, Wang TC, Yao H, Embree MC. Lgr5-expressing secretory cells form a Wnt inhibitory niche in cartilage critical for chondrocyte identity. Cell Stem Cell 2023; 30:1179-1198.e7. [PMID: 37683603 PMCID: PMC10790417 DOI: 10.1016/j.stem.2023.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 06/06/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023]
Abstract
Osteoarthritis is a degenerative joint disease that causes pain, degradation, and dysfunction. Excessive canonical Wnt signaling in osteoarthritis contributes to chondrocyte phenotypic instability and loss of cartilage homeostasis; however, the regulatory niche is unknown. Using the temporomandibular joint as a model in multiple species, we identify Lgr5-expressing secretory cells as forming a Wnt inhibitory niche that instruct Wnt-inactive chondroprogenitors to form the nascent synovial joint and regulate chondrocyte lineage and identity. Lgr5 ablation or suppression during joint development, aging, or osteoarthritis results in depletion of Wnt-inactive chondroprogenitors and a surge of Wnt-activated, phenotypically unstable chondrocytes with osteoblast-like properties. We recapitulate the cartilage niche and create StemJEL, an injectable hydrogel therapy combining hyaluronic acid and sclerostin. Local delivery of StemJEL to post-traumatic osteoarthritic jaw and knee joints in rabbit, rat, and mini-pig models restores cartilage homeostasis, chondrocyte identity, and joint function. We provide proof of principal that StemJEL preserves the chondrocyte niche and alleviates osteoarthritis.
Collapse
Affiliation(s)
- Angela Ruscitto
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peng Chen
- Clemson University-Medical University of South Carolina Joint Bioengineering Program, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ikue Tosa
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ziyi Wang
- Department of Molecular Biology and Biochemistry, Okayama University Graduate, School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 7008525, Japan
| | - Gan Zhou
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ingrid Safina
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ran Wei
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mallory M Morel
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alia Koch
- Section of Hospital Dentistry, Division of Oral & Maxillofacial Surgery, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michael Forman
- Section of Hospital Dentistry, Division of Oral & Maxillofacial Surgery, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Gwendolyn Reeve
- Division of Oral and Maxillofacial Surgery, New York Presbyterian Weill Cornell Medicine, New York, NY 10065, USA
| | - Michael K Lecholop
- Department of Oral and Maxillofacial Surgery, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Marshall Wilson
- Clemson University-Medical University of South Carolina Joint Bioengineering Program, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Daniel Bonthius
- Clemson University-Medical University of South Carolina Joint Bioengineering Program, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mo Chen
- Wnt Scientific, LLC, Harlem Biospace, New York, NY 10027, USA
| | - Mitsuaki Ono
- Department of Molecular Biology and Biochemistry, Okayama University Graduate, School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 7008525, Japan; Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 7008525, Japan
| | - Timothy C Wang
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hai Yao
- Clemson University-Medical University of South Carolina Joint Bioengineering Program, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mildred C Embree
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
17
|
Shen P, Serve S, Wu P, Liu X, Dai Y, Durán-Hernández N, Nguyen DTM, Fuchs M, Maleitzke T, Reisener MJ, Dzamukova M, Nussbaumer K, Brunner TM, Li Y, Holecska V, Heinz GA, Heinrich F, Durek P, Katsoula G, Gwinner C, Jung T, Zeggini E, Winkler T, Mashreghi MF, Pumberger M, Perka C, Löhning M. NOS inhibition reverses TLR2-induced chondrocyte dysfunction and attenuates age-related osteoarthritis. Proc Natl Acad Sci U S A 2023; 120:e2207993120. [PMID: 37428931 PMCID: PMC10629581 DOI: 10.1073/pnas.2207993120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 04/20/2023] [Indexed: 07/12/2023] Open
Abstract
Osteoarthritis (OA) is a joint disease featuring cartilage breakdown and chronic pain. Although age and joint trauma are prominently associated with OA occurrence, the trigger and signaling pathways propagating their pathogenic aspects are ill defined. Following long-term catabolic activity and traumatic cartilage breakdown, debris accumulates and can trigger Toll-like receptors (TLRs). Here we show that TLR2 stimulation suppressed the expression of matrix proteins and induced an inflammatory phenotype in human chondrocytes. Further, TLR2 stimulation impaired chondrocyte mitochondrial function, resulting in severely reduced adenosine triphosphate (ATP) production. RNA-sequencing analysis revealed that TLR2 stimulation upregulated nitric oxide synthase 2 (NOS2) expression and downregulated mitochondria function-associated genes. NOS inhibition partially restored the expression of these genes, and rescued mitochondrial function and ATP production. Correspondingly, Nos2-/- mice were protected from age-related OA development. Taken together, the TLR2-NOS axis promotes human chondrocyte dysfunction and murine OA development, and targeted interventions may provide therapeutic and preventive approaches in OA.
Collapse
Affiliation(s)
- Ping Shen
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Sciences and Technology, Xinxiang Medical University, 453003Xinxiang, China
| | - Sebastian Serve
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Peihua Wu
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Xiaohui Liu
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Yujie Dai
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Nayar Durán-Hernández
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Dan Thi Mai Nguyen
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Michael Fuchs
- Department of Orthopaedic Surgery, University of Ulm, 89081Ulm, Germany
| | - Tazio Maleitzke
- Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 13353Berlin, Germany
- Berlin Institute of Health Charité Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health at Charité–Universitätsmedizin, 10178Berlin, Germany
| | - Marie-Jacqueline Reisener
- Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Maria Dzamukova
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Katrin Nussbaumer
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
| | - Tobias M. Brunner
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Yonghai Li
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Sciences and Technology, Xinxiang Medical University, 453003Xinxiang, China
| | - Vivien Holecska
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Gitta A. Heinz
- Systems Rheumatology and Therapeutic Gene Regulation, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
| | - Frederik Heinrich
- Systems Rheumatology and Therapeutic Gene Regulation, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
| | - Pawel Durek
- Systems Rheumatology and Therapeutic Gene Regulation, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
| | - Georgia Katsoula
- Technical University of Munich School of Medicine, Technical University of Munich, Graduate School of Experimental Medicine, 81675Munich, Germany
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, 85764Neuherberg, Germany
| | - Clemens Gwinner
- Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Tobias Jung
- Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, 85764Neuherberg, Germany
- Technical University of Munich School of Medicine, Technical University of Munich and Klinikum Rechts der Isar, 81675Munich, Germany
| | - Tobias Winkler
- Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 13353Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité ‒ Universitätsmedizin Berlin, 13353Berlin, Germany
| | - Mir-Farzin Mashreghi
- Systems Rheumatology and Therapeutic Gene Regulation, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
| | - Matthias Pumberger
- Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Carsten Perka
- Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| | - Max Löhning
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center, a Leibniz Institute, 10117Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117Berlin, Germany
| |
Collapse
|
18
|
Krawetz RJ, Larijani L, Corpuz JM, Ninkovic N, Das N, Olsen A, Mohtadi N, Rezansoff A, Dufour A. Mesenchymal progenitor cells from non-inflamed versus inflamed synovium post-ACL injury present with distinct phenotypes and cartilage regeneration capacity. Stem Cell Res Ther 2023; 14:168. [PMID: 37357305 DOI: 10.1186/s13287-023-03396-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 06/05/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic debilitating disease impacting a significant percentage of the global population. While there are numerous surgical and non-invasive interventions that can postpone joint replacement, there are no current treatments which can reverse the joint damage occurring during the pathogenesis of the disease. While many groups are investigating the use of stem cell therapies in the treatment of OA, we still don't have a clear understanding of the role of these cells in the body, including heterogeneity of tissue resident adult mesenchymal progenitor cells (MPCs). METHODS In the current study, we examined MPCs from the synovium and individuals with or without a traumatic knee joint injury and explored the chondrogenic differentiation capacity of these MPCs in vitro and in vivo. RESULTS We found that there is heterogeneity of MPCs with the adult synovium and distinct sub-populations of MPCs and the abundancy of these sub-populations change with joint injury. Furthermore, only some of these sub-populations have the ability to effect cartilage repair in vivo. Using an unbiased proteomics approach, we were able to identify cell surface markers that identify this pro-chondrogenic MPC population in normal and injured joints, specifically CD82LowCD59+ synovial MPCs have robust cartilage regenerative properties in vivo. CONCLUSIONS The results of this study clearly show that cells within the adult human joint can impact cartilage repair and that these sub-populations exist within joints that have undergone a traumatic joint injury. Therefore, these populations can be exploited for the treatment of cartilage injuries and OA in future clinical trials.
Collapse
Affiliation(s)
- Roman J Krawetz
- McCaig Institute for Bone and Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
- Department Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada.
- Department of Surgery, University of Calgary, Calgary, AB, Canada.
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada.
| | - Leila Larijani
- McCaig Institute for Bone and Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Jessica May Corpuz
- McCaig Institute for Bone and Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
| | - Nicoletta Ninkovic
- McCaig Institute for Bone and Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Nabangshu Das
- McCaig Institute for Bone and Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Alexandra Olsen
- McCaig Institute for Bone and Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
| | - Nicholas Mohtadi
- McCaig Institute for Bone and Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
- Department of Surgery, University of Calgary, Calgary, AB, Canada
- Sport Medicine Centre, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Alexander Rezansoff
- McCaig Institute for Bone and Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
- Department of Surgery, University of Calgary, Calgary, AB, Canada
- Sport Medicine Centre, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- McCaig Institute for Bone and Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
19
|
Ouyang Z, Dong L, Yao F, Wang K, Chen Y, Li S, Zhou R, Zhao Y, Hu W. Cartilage-Related Collagens in Osteoarthritis and Rheumatoid Arthritis: From Pathogenesis to Therapeutics. Int J Mol Sci 2023; 24:9841. [PMID: 37372989 DOI: 10.3390/ijms24129841] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Collagens serve essential mechanical functions throughout the body, particularly in the connective tissues. In articular cartilage, collagens provide most of the biomechanical properties of the extracellular matrix essential for its function. Collagen plays a very important role in maintaining the mechanical properties of articular cartilage and the stability of the ECM. Noteworthily, many pathogenic factors in the course of osteoarthritis and rheumatoid arthritis, such as mechanical injury, inflammation, and senescence, are involved in the irreversible degradation of collagen, leading to the progressive destruction of cartilage. The degradation of collagen can generate new biochemical markers with the ability to monitor disease progression and facilitate drug development. In addition, collagen can also be used as a biomaterial with excellent properties such as low immunogenicity, biodegradability, biocompatibility, and hydrophilicity. This review not only provides a systematic description of collagen and analyzes the structural characteristics of articular cartilage and the mechanisms of cartilage damage in disease states but also provides a detailed characterization of the biomarkers of collagen production and the role of collagen in cartilage repair, providing ideas and techniques for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Ziwei Ouyang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| | - Lei Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| | - Feng Yao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Ke Wang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Shufang Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Renpeng Zhou
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| |
Collapse
|
20
|
Richard D, Capellini TD, Diekman BO. Epigenetics as a mediator of genetic risk in osteoarthritis: role during development, homeostasis, aging, and disease progression. Am J Physiol Cell Physiol 2023; 324:C1078-C1088. [PMID: 36971423 PMCID: PMC10191130 DOI: 10.1152/ajpcell.00574.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023]
Abstract
The identification of genomic loci that are associated with osteoarthritis (OA) has provided a starting point for understanding how genetic variation activates catabolic processes in the joint. However, genetic variants can only alter gene expression and cellular function when the epigenetic environment is permissive to these effects. In this review, we provide examples of how epigenetic shifts at distinct life stages can alter the risk for OA, which we posit is critical for the proper interpretation of genome-wide association studies (GWAS). During development, intensive work on the growth and differentiation factor 5 (GDF5) locus has revealed the importance of tissue-specific enhancer activity in controlling both joint development and the subsequent risk for OA. During homeostasis in adults, underlying genetic risk factors may help establish beneficial or catabolic "set points" that dictate tissue function, with a strong cumulative effect on OA risk. During aging, methylation changes and the reorganization of chromatin can "unmask" the effects of genetic variants. The destructive function of variants that alter aging would only mediate effects after reproductive competence and thus avoid any evolutionary selection pressure, as consistent with larger frameworks of biological aging and its relationship to disease. A similar "unmasking" may occur during OA progression, which is supported by the finding of distinct expression quantitative trait loci (eQTLs) in chondrocytes depending on the degree of tissue degradation. Finally, we propose that massively parallel reporter assays (MPRAs) will be a valuable tool to test the function of putative OA GWAS variants in chondrocytes from different life stages.
Collapse
Affiliation(s)
- Daniel Richard
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, United States
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Brian O Diekman
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, United States
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
21
|
Li X, Roemer FW, Cicuttini F, MacKay JW, Turmezei T, Link TM. Early knee OA definition-what do we know at this stage? An imaging perspective. Ther Adv Musculoskelet Dis 2023; 15:1759720X231158204. [PMID: 36937824 PMCID: PMC10017942 DOI: 10.1177/1759720x231158204] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/01/2023] [Indexed: 03/16/2023] Open
Abstract
While criteria for early-stage knee osteoarthritis (OA) in a primary care setting have been proposed, the role of imaging has been limited to radiography using the standard Kellgren-Lawrence classification. Standardized imaging and interpretation are critical with radiographs, yet studies have also shown that even early stages of radiographic OA already demonstrate advanced damage to knee joint tissues such as cartilage, menisci, and bone marrow. Morphological magnetic resonance imaging (MRI) shows degenerative damage earlier than radiographs and definitions for OA using MRI have been published though no accepted definition of early OA based on MRI is currently available. The clinical significance of structural abnormalities has also not been well defined, and the differentiation between normal aging and structural OA development remains a challenge. Compositional MRI of cartilage provides information on biochemical, degenerative changes within the cartilage matrix before cartilage defects occur and when cartilage damage is potentially reversible. Studies have shown that cartilage composition can predict cartilage loss and radiographic OA. However, while this technology is most promising for characterizing early OA it has currently limited clinical application. Better standardization of compositional MRI is required, which is currently work in progress. Finally, there has been renewed interest in computed tomography (CT) for assessing early knee OA as new techniques such as weight bearing and spectral CT are available, which may provide information on joint loading, cartilage, and bone and potentially have a role in better characterizing early OA. In conclusion, while imaging may have a limited role in diagnosing early OA in a primary care setting, there are advanced imaging technologies available, which detect early degeneration and may thus significantly alter management as new therapeutic modalities evolve.
Collapse
Affiliation(s)
- Xiaojuan Li
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Frank W. Roemer
- Department of Radiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Flavia Cicuttini
- Musculoskeletal Unit, Monash University and Rheumatology, Alfred Hospital, Melbourne, VIC, Australia
| | - Jamie W. MacKay
- Department of Radiology, University of Cambridge, Cambridge, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Tom Turmezei
- Department of Radiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Thomas M. Link
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 400 Parnassus Ave, A-367, San Francisco, CA 94143, USA
| |
Collapse
|
22
|
Rapp AE, Zaucke F. Cartilage extracellular matrix-derived matrikines in osteoarthritis. Am J Physiol Cell Physiol 2023; 324:C377-C394. [PMID: 36571440 DOI: 10.1152/ajpcell.00464.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Osteoarthritis (OA) is among the most frequent diseases of the musculoskeletal system. Degradation of cartilage extracellular matrix (ECM) is a hallmark of OA. During the degradation process, intact/full-length proteins and proteolytic fragments are released which then might induce different downstream responses via diverse receptors, therefore leading to different biological consequences. Collagen type II and the proteoglycan aggrecan are the most abundant components of the cartilage ECM. However, over the last decades, a large number of minor components have been identified and for some of those, a role in the manifold processes associated with OA has already been demonstrated. To date, there is still no therapy able to halt or cure OA. A better understanding of the matrikine landscape occurring with or even preceding obvious degenerative changes in joint tissues is needed and might help to identify molecules that could serve as biomarkers, druggable targets, or even be blueprints for disease modifying drug OA drugs. For this narrative review, we screened PubMed for relevant literature in the English language and summarized the current knowledge regarding the function of selected ECM molecules and the derived matrikines in the context of cartilage and OA.
Collapse
Affiliation(s)
- Anna E Rapp
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
23
|
Korcari A, Nichols AEC, Buckley MR, Loiselle AE. Scleraxis-lineage cells are required for tendon homeostasis and their depletion induces an accelerated extracellular matrix aging phenotype. eLife 2023; 12:e84194. [PMID: 36656751 PMCID: PMC9908079 DOI: 10.7554/elife.84194] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Aged tendons have disrupted homeostasis, increased injury risk, and impaired healing capacity. Understanding mechanisms of homeostatic disruption is crucial for developing therapeutics to retain tendon health through the lifespan. Here, we developed a novel model of accelerated tendon extracellular matrix (ECM) aging via depletion of Scleraxis-lineage cells in young mice (Scx-DTR). Scx-DTR recapitulates many aspects of tendon aging including comparable declines in cellularity, alterations in ECM structure, organization, and composition. Single-cell RNA sequencing demonstrated a conserved decline in tenocytes associated with ECM biosynthesis in aged and Scx-DTR tendons, identifying the requirement for Scleraxis-lineage cells during homeostasis. However, the remaining cells in aged and Scx-DTR tendons demonstrate functional divergence. Aged tenocytes become pro-inflammatory and lose proteostasis. In contrast, tenocytes from Scx-DTR tendons demonstrate enhanced remodeling capacity. Collectively, this study defines Scx-DTR as a novel model of accelerated tendon ECM aging and identifies novel biological intervention points to maintain tendon function through the lifespan.
Collapse
Affiliation(s)
- Antonion Korcari
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Anne EC Nichols
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
| | - Mark R Buckley
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| |
Collapse
|
24
|
Mechanochemistry of collagen. Acta Biomater 2023; 163:50-62. [PMID: 36669548 DOI: 10.1016/j.actbio.2023.01.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 01/02/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023]
Abstract
The collagen molecular family is the result of nearly one billion years of evolution. It is a unique family of proteins, the majority of which provide general mechanical support to biological tissues. Fibril forming collagens are the most abundant collagens in vertebrate animals and are generally found in positions that resist tensile loading. In animals, cells produce fibril-forming collagen molecules that self-assemble into larger structures known as collagen fibrils. Collagen fibrils are the fundamental, continuous, load-bearing elements in connective tissues, but are often further aggregated into larger load-bearing structures, fascicles in tendon, lamellae in cornea and in intervertebral disk. We know that failure to form fibrillar collagen is embryonic lethal, and excessive collagen formation/growth (fibrosis) or uncontrolled enzymatic remodeling (type II collagen: osteoarthritis) is pathological. Collagen is thus critical to vertebrate viability and instrumental in maintaining efficient mechanical structures. However, despite decades of research, our understanding of collagen matrix formation is not complete, and we know still less about the detailed mechanisms that drive collagen remodeling, growth, and pathology. In this perspective, we examine the known role of mechanical force on the formation and development of collagenous structure. We then discuss a mechanochemical mechanism that has the potential to unify our understanding of collagenous tissue assembly dynamics, which preferentially deposits and grows collagen fibrils directly in the path of mechanical force, where the energetics should be dissuasive and where collagen fibrils are most required. We term this mechanism: Mechanochemical force-structure causality. STATEMENT OF SIGNIFICANCE: Our mechanochemical-force structure causality postulate suggests that collagen molecules are components of mechanochemically-sensitive and dynamically-responsive fibrils. Collagen molecules assemble preferentially in the path of applied strain, can be grown in place by mechanical extension, and are retained in the path of force through strain-stabilization. The mechanisms that drive this behavior operate at the level of the molecules themselves and are encoded into the structure of the biomaterial. The concept might change our understanding of structure formation, enhance our ability to treat injuries, and accelerate the development of therapeutics to prevent pathologies such as fibrosis. We suggest that collagen is a mechanochemically responsive dynamic element designed to provide a substantial "material assist" in the construction of adaptive carriers of mechanical signals.
Collapse
|
25
|
Gialeli C, Gonçalves I. Evaluation of Glycosaminoglycans Biological Age in Cells and Tissues. Methods Mol Biol 2023; 2619:91-98. [PMID: 36662464 DOI: 10.1007/978-1-0716-2946-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Bomb pulse refers to the sharp increase of the atmospheric levels of the carbon isotope 14C due to nuclear testing during the late 1950s and early 1960s. After the nuclear test ban, atmospheric 14C levels decreased exponentially because of the diffusion and equilibration with the ocean and biosphere, as well as the incorporation indirectly into all living cells. Thus, assessing the anthropogenic 14C serves as an isotopic chronometer providing the opportunity to estimate the age of a tissue and its components with a relatively high precision comparing it with the atmospheric levels at a certain time stamp.This protocol describes the use of the bomb pulse dating as a retrospective tracer of tissue components such as the glycosaminoglycans (GAGs) and subsequent proteoglycans turnover using accelerator mass spectrometry.
Collapse
Affiliation(s)
- Chrysostomi Gialeli
- Department of Clinical Sciences Malmö, Lund University, Clinical Research Center, Malmö, Sweden.
| | - Isabel Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Clinical Research Center, Malmö, Sweden
- Department of Cardiology, Malmö, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
26
|
Davison AS, Norman BP. Alkaptonuria – Past, present and future. Adv Clin Chem 2023. [DOI: 10.1016/bs.acc.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
27
|
Wang FH, Hsieh CY, Shen CI, Chuang CH, Chung YH, Kuo CC, Lee KD, Lin CL, Su HL. Induction of type II collagen expression in M2 macrophages derived from peripheral blood mononuclear cells. Sci Rep 2022; 12:21663. [PMID: 36522405 PMCID: PMC9755523 DOI: 10.1038/s41598-022-25764-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The human type II collagen (Col II), specifically expressed in chondrocytes, is a crucial component of the adult hyaline cartilage. We examine the potential of artificial induction of Col II in human peripheral blood mononuclear cells (PBMNCs) as a novel Col II provider. Human PBMNCs were purified and were treated with high doses of macrophage-colony stimulating factor (M-CSF), granulocyte macrophage-colony stimulating factor (GM-CSF), or granulocyte-colony stimulating factor (G-CSF) and examined the Col II expression at indicated days. Quantitative Col II expression was validated by real-time reverse transcriptase-polymerase chain reaction (RT-PCR), immunocytochemistry, and flow cytometry. We demonstrate that monocytes in PBMNCs can be artificially induced to express both Col II proteins and M2 macrophage markers by the high concentration of colony-stimulating factors, especially M-CSF and GM-CSF. The Col II proteins were detected on the cell membrane and in the cytoplasm by flow cytometry and immunocytostaining. Combination with IL-4 provided a synergistic effect with M-CSF/GM-CSF to trigger Col II expression in M2 macrophages. These CD206 and Col II double-expressing cells, named modified macrophages, share M2 macrophages' anti-inflammatory potency. We demonstrated that the modified macrophages could significantly attenuate the inflammatory progress of Complete Freund's adjuvant (CFA)-induced arthritis and collagen-induced arthritis in rodents. Here, we provide the first evidence that a modified macrophage population could ectopically express Col II and control the progress of arthritis in animals.
Collapse
Affiliation(s)
- Fu-Hui Wang
- Duogenic StemCells Corporation, Taichung, Taiwan
| | | | | | - Chang-Han Chuang
- grid.452796.b0000 0004 0634 3637Department of Orthopedics, Show Chwan Memorial Hospital, Changhua, Taiwan ,grid.260542.70000 0004 0532 3749National Chung Hsing University, Taichung, Taiwan ,grid.260542.70000 0004 0532 3749Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Hsuan Chung
- grid.452796.b0000 0004 0634 3637Department of Orthopedics, Show Chwan Memorial Hospital, Changhua, Taiwan ,grid.260542.70000 0004 0532 3749National Chung Hsing University, Taichung, Taiwan ,grid.260542.70000 0004 0532 3749Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chi-Chung Kuo
- grid.414692.c0000 0004 0572 899XDepartment of Neurology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan ,grid.411824.a0000 0004 0622 7222School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| | - Kuan-Der Lee
- grid.410764.00000 0004 0573 0731Department of Medical Research and Cell Therapy and Regenerative Medicine Center, Taichung Veterans General Hospital, Taichung, Taiwan ,grid.412896.00000 0000 9337 0481Department of Medicine and Center for Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Lung Lin
- grid.252470.60000 0000 9263 9645Department of Neurosurgery, Asia University Hospital, 222, Fuxin Rd., Wufeng Dist., Taichung City, Taiwan ,grid.252470.60000 0000 9263 9645Department of Occupational Therapy, Asia University, Taichung, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, National Chung Hsing University, 145, Xin-Da Rd., South Dist., Taichung City, 402 Taiwan
| |
Collapse
|
28
|
Dynamic compression inhibits cytokine-mediated type II collagen degradation. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100292. [DOI: 10.1016/j.ocarto.2022.100292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
|
29
|
Chen P, Liu X, Gu C, Zhong P, Song N, Li M, Dai Z, Fang X, Liu Z, Zhang J, Tang R, Fan S, Lin X. A plant-derived natural photosynthetic system for improving cell anabolism. Nature 2022; 612:546-554. [PMID: 36477541 PMCID: PMC9750875 DOI: 10.1038/s41586-022-05499-y] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 10/31/2022] [Indexed: 12/12/2022]
Abstract
Insufficient intracellular anabolism is a crucial factor involved in many pathological processes in the body1,2. The anabolism of intracellular substances requires the consumption of sufficient intracellular energy and the production of reducing equivalents. ATP acts as an 'energy currency' for biological processes in cells3,4, and the reduced form of NADPH is a key electron donor that provides reducing power for anabolism5. Under pathological conditions, it is difficult to correct impaired anabolism and to increase insufficient levels of ATP and NADPH to optimum concentrations1,4,6-8. Here we develop an independent and controllable nanosized plant-derived photosynthetic system based on nanothylakoid units (NTUs). To enable cross-species applications, we use a specific mature cell membrane (the chondrocyte membrane (CM)) for camouflage encapsulation. As proof of concept, we demonstrate that these CM-NTUs enter chondrocytes through membrane fusion, avoid lysosome degradation and achieve rapid penetration. Moreover, the CM-NTUs increase intracellular ATP and NADPH levels in situ following exposure to light and improve anabolism in degenerated chondrocytes. They can also systemically correct energy imbalance and restore cellular metabolism to improve cartilage homeostasis and protect against pathological progression of osteoarthritis. Our therapeutic strategy for degenerative diseases is based on a natural photosynthetic system that can controllably enhance cell anabolism by independently providing key energy and metabolic carriers. This study also provides an enhanced understanding of the preparation and application of bioorganisms and composite biomaterials for the treatment of disease.
Collapse
Affiliation(s)
- Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xin Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Chenhui Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Peiyu Zhong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Nan Song
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Mobai Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhanqiu Dai
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangqian Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhaoming Liu
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Jianfeng Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, China.
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
30
|
Otarola GA, Hu JC, Athanasiou KA. Ion modulatory treatments toward functional self-assembled neocartilage. Acta Biomater 2022; 153:85-96. [PMID: 36113725 PMCID: PMC11575480 DOI: 10.1016/j.actbio.2022.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022]
Abstract
Signals that recapitulate in vitro the conditions found in vivo, such as hypoxia or mechanical forces, contribute to the generation of tissue-engineered hyaline-like tissues. The cell regulatory processes behind hypoxic and mechanical stimuli rely on ion concentration; iron is required to degrade the hypoxia inducible factor 1a (HIF1α) under normoxia, whereas the initiation of mechanotransduction requires the cytoplasmic increase of calcium concentration. In this work, we propose that ion modulation can be used to improve the biomechanical properties of self-assembled neocartilage constructs derived from rejuvenated expanded minipig rib chondrocytes. The objectives of this work were 1) to determine the effects of iron sequestration on self-assembled neocartilage constructs using two doses of the iron chelator deferoxamine (DFO), and 2) to evaluate the performance of the combined treatment of DFO and ionomycin, a calcium ionophore that triggers cytoplasmic calcium accumulation. This study employed a two-phase approach. In Phase I, constructs treated with a high dose of DFO (100 µM) exhibited an 87% increase in pyridinoline crosslinks, a 57% increase in the Young's modulus, and a 112% increase in the ultimate tensile strength (UTS) of the neotissue. In Phase II, the combined use of both ion modulators resulted in 150% and 176% significant increases in the Young's modulus and UTS of neocartilage constructs, respectively; for the first time, neocartilage constructs achieved a Young's modulus of 11.76±3.29 MPa and UTS of 4.20±1.24 MPa. The results of this work provide evidence that ion modulation can be employed to improve the biomechanical properties in engineered neotissues. STATEMENT OF SIGNIFICANCE: The translation of tissue-engineered products requires the development of strategies capable of producing biomimetic neotissues in a replicable, controllable, and cost-effective manner. Among other functions, Fe2+ and Ca2+ are involved in the control of the hypoxic response and mechanotransduction, respectively. Both stimuli, hypoxia and mechanical forces, are known to favor chondrogenesis. This study utilized ion modulators to improve the mechanical properties self-assembled neocartilage constructs derived from expanded and rejuvenated costal chondrocytes via Fe2+ sequestration and Ca2+ influx, alone or in combination. The results indicate that ion modulation induced tissue maturation and a significant improvement of the mechanical properties, and holds potential as a tool to mitigate the need for bioreactors and engineer hyaline-like tissues.
Collapse
Affiliation(s)
- Gaston A Otarola
- 3131 Engineering Hall, Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
| | - Jerry C Hu
- 3131 Engineering Hall, Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
| | - Kyriacos A Athanasiou
- 3131 Engineering Hall, Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA.
| |
Collapse
|
31
|
Evans LAE, Pitsillides AA. Structural clues to articular calcified cartilage function: A descriptive review of this crucial interface tissue. J Anat 2022; 241:875-895. [PMID: 35866709 PMCID: PMC9482704 DOI: 10.1111/joa.13728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 11/26/2022] Open
Abstract
Articular calcified cartilage (ACC) has been dismissed, by some, as a remnant of endochondral ossification without functional relevance to joint articulation or weight-bearing. Recent research indicates that morphologic and metabolic ACC features may be important, reflecting knee joint osteoarthritis (OA) predisposition. ACC is less investigated than neighbouring joint tissues, with its component chondrocytes and mineralised matrix often being either ignored or integrated into analyses of hyaline articular cartilage and subchondral bone tissue respectively. Anatomical variation in ACC is recognised between species, individuals and age groups, but the selective pressures underlying this variation are unknown. Consequently, optimal ACC biomechanical features are also unknown as are any potential locomotory roles. This review collates descriptions of ACC anatomy and biology in health and disease, with a view to revealing its structure/function relationship and highlighting potential future research avenues. Mouse models of healthy and OA joint ageing have shown disparities in ACC load-induced deformations at the knee joint. This raises the hypothesis that ACC response to locomotor forces over time may influence, or even underlie, the bony and hyaline cartilage symptoms characteristic of OA. To effectively investigate the ACC, greater resolution of joint imaging and merging of hierarchical scale data will be required. An appreciation of OA as a 'whole joint disease' is expanding, as is the possibility that the ACC may be a key player in healthy ageing and in the transition to OA joint pathology.
Collapse
Affiliation(s)
- Lucinda A. E. Evans
- Department of Comparative Biomedical SciencesRoyal Veterinary College, University of LondonLondonUK
| | - Andrew A. Pitsillides
- Department of Comparative Biomedical SciencesRoyal Veterinary College, University of LondonLondonUK
| |
Collapse
|
32
|
Khanijou JK, Yee Z, Raida M, Lee JM, Tay EZE, Gruber J, Walczyk T. Efficiency of Protein Renewal Is Limited by Feed Intake and Not by Protein Lifetime in Aging Caenorhabditis elegans. J Proteome Res 2022; 21:2664-2686. [PMID: 36181456 DOI: 10.1021/acs.jproteome.2c00383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein turnover maintains the proteome's functional integrity. Here, protein turnover efficiency over time in wild-type Caenorhabditis elegans was assessed using inverse [15N]-pulse labeling up to 7 days after the egg-laying phase at 20 °C. Isotopic analysis of some abundant proteins was executed favoring data quality over quantity for mathematical modeling. Surprisingly, isotopic enrichment over time reached an upper limit showing an apparent cessation of protein renewal well before death, with protein fractions inaccessible to turnover ranging from 14 to 83%. For life span modulation, worms were raised at different temperatures after egg laying. Mathematical modeling of isotopic enrichment points either to a slowdown of protein turnover or to an increasing protein fraction resistant to turnover with time. Most notably, the estimated time points of protein turnover cessation from our mathematical model were highly correlated with the observed median life span. Thrashing and pumping rates over time were linearly correlated with isotopic enrichment, therefore linking protein/tracer intake to protein turnover rate and protein life span. If confirmed, life span extension is possible by optimizing protein turnover rate through modulating protein intake in C. elegans and possibly other organisms. While proteome maintenance benefits from a high protein turnover rate, protein turnover is fundamentally energy-intensive, where oxidative stress contributes to damage that it is supposed to repair.
Collapse
Affiliation(s)
- Jasmeet Kaur Khanijou
- Department of Chemistry, National University of Singapore (NUS), Singapore 117543, Singapore.,Shared Analytics, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
| | - Zhuangli Yee
- Yale-NUS College, Singapore 138527, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
| | - Manfred Raida
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore.,Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - Jin Meng Lee
- Department of Chemistry, National University of Singapore (NUS), Singapore 117543, Singapore
| | - Evan Zhi En Tay
- Department of Chemistry, National University of Singapore (NUS), Singapore 117543, Singapore
| | - Jan Gruber
- Yale-NUS College, Singapore 138527, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
| | - Thomas Walczyk
- Department of Chemistry, National University of Singapore (NUS), Singapore 117543, Singapore
| |
Collapse
|
33
|
Hines MR, Goetz JE, Gomez-Contreras PC, Rodman SN, Liman S, Femino EL, Kluz PN, Wagner BA, Buettner GR, Kelley EE, Coleman MC. Extracellular biomolecular free radical formation during injury. Free Radic Biol Med 2022; 188:175-184. [PMID: 35724853 PMCID: PMC9725094 DOI: 10.1016/j.freeradbiomed.2022.06.223] [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: 03/25/2022] [Revised: 05/15/2022] [Accepted: 06/11/2022] [Indexed: 11/30/2022]
Abstract
Determine if oxidative damage increases in articular cartilage as a result of injury and matrix failure and whether modulation of the local redox environment influences this damage. Osteoarthritis is an age associated disease with no current disease modifying approaches available. Mechanisms of cartilage damage in vitro suggest tissue free radical production could be critical to early degeneration, but these mechanisms have not been described in intact tissue. To assess free radical production as a result of traumatic injury, we measured biomolecular free radical generation via immuno-spin trapping (IST) of protein/proteoglycan/lipid free radicals after a 2 J/cm2 impact to swine articular cartilage explants. This technique allows visualization of free radical formation upon a wide variety of molecules using formalin-fixed, paraffin-embedded approaches. Scoring of extracellular staining by trained, blinded scorers demonstrated significant increases with impact injury, particularly at sites of cartilage cracking. Increases remain in the absence of live chondrocytes but are diminished; thus, they appear to be a cell-dependent and -independent feature of injury. We then modulated the extracellular environment with a pulse of heparin to demonstrate the responsiveness of the IST signal to changes in cartilage biology. Addition of heparin caused a distinct change in the distribution of protein/lipid free radicals at sites of failure alongside a variety of pertinent redox changes related to osteoarthritis. This study directly confirms the production of biomolecular free radicals from articular trauma, providing a rigorous characterization of their formation by injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Paige N Kluz
- University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | | |
Collapse
|
34
|
Olesen AT, Malchow-Møller L, Bendixen RD, Kjær M, Mackey AL, Magnusson SP, Svensson RB. Intramuscular connective tissue content and mechanical properties: Influence of aging and physical activity in mice. Exp Gerontol 2022; 166:111893. [PMID: 35870752 DOI: 10.1016/j.exger.2022.111893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/05/2022] [Accepted: 07/09/2022] [Indexed: 11/25/2022]
Abstract
Aging is accompanied by morphological and mechanical changes to the intramuscular connective tissue (IMCT) of skeletal muscles, but whether physical exercise can influence these changes is debated. We investigated the effects of aging and exercise with high or low resistance on composition and mechanical properties of the IMCT, including direct measurements on isolated IMCT which has rarely been reported. Middle-aged (11 months, n = 24) and old (22 months, n = 18) C57BL/6 mice completed either high (HR) or low (LR) resistance voluntary wheel running or were sedentary (SED) for 10 weeks. Passive mechanical properties of the intact soleus and plantaris muscles and the isolated IMCT of the plantaris muscle were measured in vitro. IMCT thickness was measured on picrosirius red stained cross sections of the gastrocnemius and soleus muscle and for the gastrocnemius hydroxyproline content was quantified biochemically and advanced glycation end-products (AGEs) estimated by fluorometry. Mechanical stiffness, IMCT content and total AGEs were all elevated with aging in agreement with previous findings but were largely unaffected by training. Conclusion: IMCT accumulated with aging with a proportional increase in mechanical stiffness, but even the relatively high exercise volume achieved with voluntary wheel-running with or without resistance did not significantly influence these changes.
Collapse
Affiliation(s)
- Annesofie T Olesen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Lasse Malchow-Møller
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Rune D Bendixen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Michael Kjær
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Denmark; XLab, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - S Peter Magnusson
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Denmark; Department of Physical and Occupational Therapy, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark
| | - Rene B Svensson
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Denmark.
| |
Collapse
|
35
|
Merrild NG, Holzmann V, Ariosa-Morejon Y, Faull PA, Coleman J, Barrell WB, Young G, Fischer R, Kelly DJ, Addison O, Vincent TL, Grigoriadis AE, Gentleman E. Local depletion of proteoglycans mediates cartilage tissue repair in an ex vivo integration model. Acta Biomater 2022; 149:179-188. [PMID: 35779773 DOI: 10.1016/j.actbio.2022.06.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/25/2022] [Accepted: 06/17/2022] [Indexed: 11/28/2022]
Abstract
Successfully replacing damaged cartilage with tissue-engineered constructs requires integration with the host tissue and could benefit from leveraging the native tissue's intrinsic healing capacity; however, efforts are limited by a poor understanding of how cartilage repairs minor defects. Here, we investigated the conditions that foster natural cartilage tissue repair to identify strategies that might be exploited to enhance the integration of engineered/grafted cartilage with host tissue. We damaged porcine articular cartilage explants and using a combination of pulsed SILAC-based proteomics, ultrastructural imaging, and catabolic enzyme blocking strategies reveal that integration of damaged cartilage surfaces is not driven by neo-matrix synthesis, but rather local depletion of proteoglycans. ADAMTS4 expression and activity are upregulated in injured cartilage explants, but integration could be reduced by inhibiting metalloproteinase activity with TIMP3. These observations suggest that catabolic enzyme-mediated proteoglycan depletion likely allows existing collagen fibrils to undergo cross-linking, fibrillogenesis, or entanglement, driving integration. Catabolic enzymes are often considered pathophysiological markers of osteoarthritis. Our findings suggest that damage-induced upregulation of metalloproteinase activity may be a part of a healing response that tips towards tissue destruction under pathological conditions and in osteoarthritis, but could also be harnessed in tissue engineering strategies to mediate repair. STATEMENT OF SIGNIFICANCE: Cartilage tissue engineering strategies require graft integration with the surrounding tissue; however, how the native tissue repairs minor injuries is poorly understood. We applied pulsed SILAC-based proteomics, ultrastructural imaging, and catabolic enzyme blocking strategies to a porcine cartilage explant model and found that integration of damaged cartilage surfaces is driven by catabolic enzyme-mediated local depletion of proteoglycans. Although catabolic enzymes have been implicated in cartilage destruction in osteoarthritis, our findings suggest that damage-induced upregulation of metalloproteinase activity may be a part of a healing response that tips towards tissue destruction under pathological conditions. They also suggest that this natural cartilage tissue repair process could be harnessed in tissue engineering strategies to enhance the integration of engineered cartilage with host tissue.
Collapse
Affiliation(s)
- Nicholas Groth Merrild
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Viktoria Holzmann
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Yoanna Ariosa-Morejon
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Peter A Faull
- College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Jennifer Coleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - William B Barrell
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Gloria Young
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Owen Addison
- Centre for Oral, Clinical and Translational Sciences, King's College London, London SE1 9RT, UK
| | - Tonia L Vincent
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | | | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK.
| |
Collapse
|
36
|
The regional turnover of cartilage collagen matrix in late-stage human knee osteoarthritis. Osteoarthritis Cartilage 2022; 30:886-895. [PMID: 35358700 DOI: 10.1016/j.joca.2022.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/03/2022] [Accepted: 03/22/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Cartilage collagen has very limited repair potential, though some turnover and incorporation has not been fully excluded. We aim to determine the regional turnover of human osteoarthritis cartilage. DESIGN Patients scheduled for knee joint replacement surgery due to osteoarthritis were recruited in this prospective study of four weeks duration. Deuterium oxide (D2O) was administered orally by weekly boluses at 70% D2O, initially 150 ml followed by three boluses of 50 ml. Cartilage from the medial tibia plateau was sampled centrally, under the meniscus, and from osteophytes and treated enzymatically with hyaluronidase and trypsin. Samples were analysed for deuterium incorporation in alanine using mass spectrometry and for gene expression by real-time reverse transcriptase polymerase chain reaction. RESULTS Twenty participants completed the study: mean (SD) age 64 ± 9.1 years, 45% female, BMI 29.5 ± 4.8 kg/m2. Enzymatically treated cartilage from central and submeniscal regions showed similar enrichments at 0.063% APE, while osteophytes showed significantly greater enrichment at 0.072% APE (95% confidence interval of difference) [0.004-0.015]). Fractional synthesis rates were similar for central 0.027%/day and submeniscal cartilage 0.022%/day but 10-fold higher in osteophytes 0.22%/day [0.098-0.363]. When compared to central cartilage, submeniscal cartilage had increased gene expression of MMP-3 and decreased lubricin expression. Untreated cartilage had higher turnover (enrichments at 0.073% APE) than enzymatically treated cartilage (0.063% APE). CONCLUSIONS In OA, despite regional differences in gene expression, the turnover of the articular cartilage matrix across the entire joint surface is very limited, but higher turnover was observed in osteophyte cartilage.
Collapse
|
37
|
Vincent TL, McClurg O, Troeberg L. The Extracellular Matrix of Articular Cartilage Controls the Bioavailability of Pericellular Matrix-Bound Growth Factors to Drive Tissue Homeostasis and Repair. Int J Mol Sci 2022; 23:6003. [PMID: 35682681 PMCID: PMC9181404 DOI: 10.3390/ijms23116003] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 11/24/2022] Open
Abstract
The extracellular matrix (ECM) has long been regarded as a packing material; supporting cells within the tissue and providing tensile strength and protection from mechanical stress. There is little surprise when one considers the dynamic nature of many of the individual proteins that contribute to the ECM, that we are beginning to appreciate a more nuanced role for the ECM in tissue homeostasis and disease. Articular cartilage is adapted to be able to perceive and respond to mechanical load. Indeed, physiological loads are essential to maintain cartilage thickness in a healthy joint and excessive mechanical stress is associated with the breakdown of the matrix that is seen in osteoarthritis (OA). Although the trigger by which increased mechanical stress drives catabolic pathways remains unknown, one mechanism by which cartilage responds to increased compressive load is by the release of growth factors that are sequestered in the pericellular matrix. These are heparan sulfate-bound growth factors that appear to be largely chondroprotective and displaced by an aggrecan-dependent sodium flux. Emerging evidence suggests that the released growth factors act in a coordinated fashion to drive cartilage repair. Thus, we are beginning to appreciate that the ECM is the key mechano-sensor and mechano-effector in cartilage, responsible for directing subsequent cellular events of relevance to joint health and disease.
Collapse
Affiliation(s)
- Tonia L. Vincent
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Oliver McClurg
- Norwich Medical School, University of East Anglia, Norwich, Norwich NR4 7UQ, UK; (O.M.); (L.T.)
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Norwich, Norwich NR4 7UQ, UK; (O.M.); (L.T.)
| |
Collapse
|
38
|
Vincent TL, Alliston T, Kapoor M, Loeser RF, Troeberg L, Little CB. Osteoarthritis Pathophysiology: Therapeutic Target Discovery may Require a Multifaceted Approach. Clin Geriatr Med 2022; 38:193-219. [PMID: 35410676 PMCID: PMC9107912 DOI: 10.1016/j.cger.2021.11.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Molecular understanding of osteoarthritis (OA) has greatly increased through careful analysis of tissue samples, preclinical models, and large-scale agnostic "-omic" studies. There is broad acceptance that systemic and biomechanical signals affect multiple tissues of the joint, each of which could potentially be targeted to improve patient outcomes. In this review six experts in different aspects of OA pathogenesis provide their independent view on what they believe to be good tractable approaches to OA target discovery. We conclude that molecular discovery has been high but future transformative studies require a multidisciplinary holistic approach to develop therapeutic strategies with high clinical efficacy.
Collapse
Affiliation(s)
- Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mohit Kapoor
- Department of Surgery and Laboratory Medicine and Pathobiology, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, University of Toronto, Toronto, Canada
| | - Richard F Loeser
- Department of Medicine, Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Linda Troeberg
- University of East Anglia, Norwich Medical School, Norwich NR4 7UQ, UK
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute University of Sydney Faculty of Medicine and Health at Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| |
Collapse
|
39
|
Bielajew BJ, Donahue RP, Lamkin EK, Hu JC, Hascall VC, Athanasiou KA. Proteomic, mechanical, and biochemical characterization of cartilage development. Acta Biomater 2022; 143:52-62. [PMID: 35235865 DOI: 10.1016/j.actbio.2022.02.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/04/2022] [Accepted: 02/23/2022] [Indexed: 01/06/2023]
Abstract
The objective of this work is to examine the development of porcine cartilage by analyzing its mechanical properties, biochemical content, and proteomics at different developmental stages. Cartilage from the knees of fetal, neonatal, juvenile, and mature pigs was analyzed using histology, mechanical testing, biochemical assays, fluorophore-assisted carbohydrate electrophoresis, and bottom-up proteomics. Mature cartilage has 2.2-times the collagen per dry weight of fetal cartilage, and fetal cartilage has 2.1-times and 17.9-times the glycosaminoglycan and DNA per dry weight of mature cartilage, respectively. Tensile and compressive properties peak in the juvenile stage, with a tensile modulus 4.7-times that of neonatal. Proteomics analysis reveals increases in collagen types II and III, while collagen types IX, XI, and XIV, and aggrecan decrease with age. For example, collagen types IX and XI decrease 9.4-times and 5.1-times, respectively from fetal to mature. Mechanical and biochemical measurements have their greatest developmental changes between the neonatal and juvenile stages, where mechanotransduction plays a major role. Bottom-up proteomics serves as a powerful tool for tissue characterization, showing results beyond those of routine biochemical analysis. For example, proteomic analysis shows significant drops in collagen types IX, XI, and XIV throughout development, which shows insight into the permanence of cartilage's matrix. Changes in overall glycosaminoglycan content compared to aggrecan and link protein indicate non-enzymatic degradation of aggrecan structures or hyaluronan in mature cartilage. In addition to tissue characterization, bottom-up proteomics techniques are critical in tissue engineering efforts toward repair or regeneration of cartilage in animal models. STATEMENT OF SIGNIFICANCE: In this study, the development of porcine articular cartilage is interrogated through biomechanical, biochemical, and proteomic techniques, to determine how mechanics and extracellular matrix composition change from fetal to mature cartilage. For the first time, a bottom-up proteomics approach is used to reveal a wide variety of protein changes through aging; for example, the collagen subtype composition of the cartilage increases in collagen types II and III, and decreases in collagen types IX, XI, and XIV. This analysis shows that bottom-up proteomics is a critical tool in tissue characterization, especially toward developing a deeper understanding of matrix composition and development in tissue engineering studies.
Collapse
|
40
|
Batool S, Hammami M, Mantebea H, Badar F, Xia Y. Location-Specific Study of Young Rabbit Femoral Cartilage by Quantitative µMRI and Polarized Light Microscopy. Cartilage 2022; 13:19476035221085143. [PMID: 35306861 PMCID: PMC9137317 DOI: 10.1177/19476035221085143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Microscopic magnetic resonance imaging (µMRI) and polarized light microscopy (PLM) are used to characterize the structural variations at different anatomical locations of femoral cartilage in young rabbits (12-14 weeks old). DESIGN Four intact knees were imaged by µMRI at 86 µm resolution. Three small cartilage-bone specimens were harvested from each of 2 femoral medial condyles and imaged by quantitative µMRI (T2 anisotropy) at 9.75 µm resolution (N = 6). These specimens, as well as the other 2 intact femoral condyles, were used for histology and imaged by quantitative PLM (retardation and angle) at 0.25 µm to 4 µm resolutions. RESULTS Quantitative MRI relaxation data and PLM fibril data revealed collaboratively distinct topographical variations in both cartilage thickness and its collagen organization in the juvenile joint. Cartilage characteristics from the central location have a 3-zone arcade-like fibril structure and a distinct magic angle effect, commonly seen in mature articular cartilage, while cartilage at the anterior location lacks these characteristics. Overall, the lowest retardation values and isotropic T2 values have been found in the distal femur (trochlear ridge), with predominant parallel fibers with respect to the articular surface. Central cartilage is the thickest (~550 µm), approximately twice as thick as the anterior and posterior locations. CONCLUSION Distinctly different characteristics of tissue properties were found in cartilage at different topographical locations on femoral condyle in rabbits. Knowledge of location-specific structural differences in the collagen network over the joint surface can improve the understanding of local mechanobiology and provide insights to tissue engineering and degradation repairs.
Collapse
Affiliation(s)
- Syeda Batool
- Department of Physics, Center for Biomedical Research, Oakland University, Rochester, MI, USA
| | - Mouhamad Hammami
- Department of Physics, Center for Biomedical Research, Oakland University, Rochester, MI, USA
| | - Hannah Mantebea
- Department of Physics, Center for Biomedical Research, Oakland University, Rochester, MI, USA
| | - Farid Badar
- Department of Physics, Center for Biomedical Research, Oakland University, Rochester, MI, USA
| | - Yang Xia
- Department of Physics, Center for Biomedical Research, Oakland University, Rochester, MI, USA,Yang Xia, Department of Physics, Center for Biomedical Research, Oakland University, 244 Meadow Brook Road, Rochester, MI 48309, USA.
| |
Collapse
|
41
|
Zoetebier B, Schmitz T, Ito K, Karperien M, Tryfonidou MA, Paez J. Injectable hydrogels for articular cartilage and nucleus pulposus repair: Status quo and prospects. Tissue Eng Part A 2022; 28:478-499. [PMID: 35232245 DOI: 10.1089/ten.tea.2021.0226] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) and chronic low back pain due to degenerative (intervertebral) disc disease (DDD) are two of the major causes of disabilities worldwide, affecting hundreds of millions of people and leading to a high socioeconomic burden. Although OA occurs in synovial joints and DDD occurs in cartilaginous joints, the similarities are striking, with both joints showing commonalities in the nature of the tissues and in the degenerative processes during disease. Consequently, repair strategies for articular cartilage (AC) and nucleus pulposus (NP), the core of the intervertebral disc, in the context of OA and DDD share common aspects. One of such tissue engineering approaches is the use of injectable hydrogels for AC and NP repair. In this review, the state-of-the-art and recent developments in injectable hydrogels for repairing, restoring, and regenerating AC tissue suffering from OA and NP tissue in DDD are summarized focusing on cell-free approaches. The various biomaterial strategies exploited for repair of both tissues are compared, and the synergies that could be gained by translating experiences from one tissue to the other are identified.
Collapse
Affiliation(s)
- Bram Zoetebier
- University of Twente Faculty of Science and Technology, 207105, Developmental BioEngineering , Drienerlolaan 5, Enschede, Netherlands, 7500 AE;
| | - Tara Schmitz
- Eindhoven University of Technology, 3169, Department of Biomedical Engineering, Eindhoven, Noord-Brabant, Netherlands;
| | - Keita Ito
- Eindhoven University of Technology, Department of Biomedical Engineering, P.O. Box 513, GEMZ 4.115, Eindhoven, Netherlands, 5600 MB;
| | | | - Marianna A Tryfonidou
- Utrecht University, Faculty of Veterinary Medicine, Clinical Sciences of Companion Animals, Yalelaan 108, Utrecht, Netherlands, 3584 CM;
| | - Julieta Paez
- University of Twente Faculty of Science and Technology, 207105, Developmental Bioengineering, University of Twente P.O. Box 217, Enschede The Netherlands, Enschede, Netherlands, 7500 AE;
| |
Collapse
|
42
|
Kobak KA, Batushansky A, Borowik AK, Lopes EPB, Peelor III FF, Donovan EL, Kinter MT, Miller BF, Griffin TM. An In Vivo Stable Isotope Labeling Method to Investigate Individual Matrix Protein Synthesis, Ribosomal Biogenesis, and Cellular Proliferation in Murine Articular Cartilage. FUNCTION 2022; 3:zqac008. [PMID: 35399495 PMCID: PMC8991031 DOI: 10.1093/function/zqac008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Targeting chondrocyte dynamics is a strategy for slowing osteoarthritis progression during aging. We describe a stable-isotope method using in vivo deuterium oxide labeling and mass spectrometry to measure protein concentration, protein half-life, cell proliferation, and ribosomal biogenesis in a single sample of murine articular cartilage. We hypothesized that a 60-d labeling period would capture age-related declines in cartilage matrix protein content, protein synthesis rates, and cellular proliferation. Knee cartilage was harvested to the subchondral bone from 25- to 90-wk-old female C57BL/6J mice treated with deuterium oxide for 15, 30, 45, and 60 d. We measured protein concentration and half-lives using targeted high resolution accurate mass spectrometry and d2ome data processing software. Deuterium enrichment was quantified in isolated DNA and RNA to measure cell proliferation and ribosomal biogenesis, respectively. Most collagen isoforms were less abundant in aged animals, with negligible collagen synthesis at either age. In contrast, age altered the concentration and half-lives of many proteoglycans and other matrix proteins, including several with greater concentration and half-lives in older mice such as proteoglycan 4, clusterin, and fibronectin-1. Cellular proteins were less abundant in older animals, consistent with reduced cellularity. Nevertheless, deuterium was maximally incorporated into 60% of DNA and RNA by 15 d of labeling in both age groups, suggesting the presence of two large pools of either rapidly (<15 d) or slowly (>60 d) proliferating cells. Our findings indicate that age-associated changes in cartilage matrix protein content and synthesis occur without detectable changes in the relative number of proliferating cells.
Collapse
Affiliation(s)
- Kamil A Kobak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw 50-367, Poland
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Erika Prado Barboza Lopes
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Frederick F Peelor III
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Elise L Donovan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Michael T Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Research & Development, Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
43
|
Shajib MS, Futrega K, Jacob Klein T, Crawford RW, Doran MR. Collagenase treatment appears to improve cartilage tissue integration but damage to collagen networks is likely permanent. J Tissue Eng 2022; 13:20417314221074207. [PMID: 35096364 PMCID: PMC8793122 DOI: 10.1177/20417314221074207] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/03/2022] [Indexed: 11/16/2022] Open
Abstract
When repairing cartilage defects a major challenge is achieving high-quality integration between the repair tissue and adjacent native cartilage. Matrix-rich cartilage is not easily remodeled, motivating several studies to trial enzyme treatment of the tissue interface to facilitate remodeling and integration. Studying and optimizing such processes is tedious, as well as potentially expensive, and thus simpler models are needed to evaluate the merits of enzyme treatment on cartilage tissue integration. Herein, we used engineered cartilage microtissues formed from bone marrow-derived stromal cells (BMSC) or expanded articular chondrocytes (ACh) to study the impact of enzyme treatment on cartilage tissue integration and matrix remodeling. A 5-min treatment with collagenase appeared to improve cartilage microtissue integration, while up to 48 h treatment with hyaluronidase did not. Alcian blue and anti-collagen II staining suggested that collagenase treatment did facilitate near seamless integration of cartilage microtissues. Microtissue sections were stained with Picrosirius red and characterized using polarized light microscopy, revealing that individual microtissues contained a collagen network organized in concentric shells. While collagenase treatment appeared to improve tissue integration, assessment of the collagen fibers with polarized light indicated that enzymatically damaged networks were not remodeled nor restored during subsequent culture. This model and these data paradoxically suggest that collagen network disruption is required to improve cartilage tissue integration, but that the disrupted collagen networks are unlikely to subsequently be restored. Future studies should attempt to limit collagen network disruption to the surface of the cartilage, and we recommend using Picrosirius red staining and polarized light to assess the quality of matrix remodeling and integration.
Collapse
Affiliation(s)
- Md. Shafiullah Shajib
- School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- Centre for Biomedical Technologies, School of Mechanical, Medical, and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Translational Research Institute, Brisbane, QLD, Australia
| | - Kathryn Futrega
- Centre for Biomedical Technologies, School of Mechanical, Medical, and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Translational Research Institute, Brisbane, QLD, Australia
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Travis Jacob Klein
- Centre for Biomedical Technologies, School of Mechanical, Medical, and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ross W Crawford
- Centre for Biomedical Technologies, School of Mechanical, Medical, and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Michael Robert Doran
- School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- Centre for Biomedical Technologies, School of Mechanical, Medical, and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Translational Research Institute, Brisbane, QLD, Australia
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Mater Research Institute – University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
44
|
Bay-Jensen AC, Mobasheri A, Thudium CS, Kraus VB, Karsdal MA. Blood and urine biomarkers in osteoarthritis - an update on cartilage associated type II collagen and aggrecan markers. Curr Opin Rheumatol 2022; 34:54-60. [PMID: 34652292 PMCID: PMC8635261 DOI: 10.1097/bor.0000000000000845] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA) is a painful disease for which drug development has proven difficult. One major reason for this is the heterogeneity of the disease and the current lack of operationalized means to distinguish various disease endotypes (molecular subtypes). Biomarkers measured in blood or urine, reflecting joint tissue turnover, have been developed and tested during the last decades. In this narrative review, we provide highlights on biomarkers derived from the two most studied and abundant cartilage proteins - type II collagen and aggrecan. RECENT FINDINGS Multiple biomarkers assessing type II collagen degradation and formation, and aggrecan turnover have been developed. Several markers, such as uCTX-II, have been validated for their association with disease severity and prognosis, as well as pharmacodynamically used to describe the mode of action and efficacy of drugs in development. There is a great need for biomarkers for subdividing patients (i.e., endotyping) and recent scientific advances have not yet come closer to achieving this goal. SUMMARY There is strong support for using biomarkers for understanding OA, reflecting degradation and formation of the joint tissues, focused on type II collagen and aggrecan. There is still a lack of in vitro diagnostics, in all contexts of use.
Collapse
Affiliation(s)
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- University Medical Center Utrecht, Department of Orthopedics, Rheumatology and Clinical Immunology, Utrecht, The Netherlands
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, University of Liege, Liege, Belgium
| | | | - Virginia B. Kraus
- Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | |
Collapse
|
45
|
Jørgensen AE, Schjerling P, Krogsgaard MR, Petersen MM, Olsen J, Kjær M, Heinemeier KM. Collagen Growth Pattern in Human Articular Cartilage of the Knee. Cartilage 2021; 13:408S-418S. [PMID: 33147986 PMCID: PMC8804751 DOI: 10.1177/1947603520971016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE During skeletal growth, the articular cartilage expands to maintain its cover of bones in joints, however, it is unclear when and how cartilage grows. We aim to determine the expanding growth pattern and timing across the tibia plateau in human knees. DESIGN Six human tibia plateaus (2 healthy, 2 with osteoarthritis, and 2 with posttraumatic osteoarthritis) were used for full-depth cartilage sampling systematically across the joint surface at 12 medial and 4 lateral sites. Methodologically, we took advantage of the performed nuclear bomb tests in the years 1955 to 1963, which increased the atmospheric 14C that was incorporated into human tissues. Cartilage was treated enzymatically to extract collagen, analyzed for 14C content, and year at formation was determined from historical atmospheric 14C concentrations. RESULTS By age-determination, each tibia condyle had central points of formation surrounded by later-formed cartilage toward the periphery. Furthermore, the tibia plateaus contained collagen with 14C levels corresponding to mean donor age of 11.7 years (±3.8 SD). Finally, the medial condyle had lower 14C levels corresponding to formation 1 year later than the lateral condyle (P = 0.009). CONCLUSIONS Human cartilage on the tibia plateau contains collagen that has experienced little if any turnover since school-age. The cartilage formation develops from 2 condyle centers and radially outward with the medial condyle finishing slightly later than the lateral condyle. This suggests a childhood programmed cartilage formation with a very limited adulthood collagen turnover.
Collapse
Affiliation(s)
- Adam E.M. Jørgensen
- Institute of Sports Medicine Copenhagen,
Department of Orthopedic Surgery M81, Bispebjerg and Frederiksberg Hospital,
Copenhagen, Denmark,Center for Healthy Aging, Faculty of
Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark,Adam E.M. Jørgensen, Institute of Sports
Medicine Copenhagen, Department of Orthopedic Surgery M81, Bispebjerg and
Frederiksberg Hospital, Nielsine Nielsensvej 11, Copenhagen, Denmark, DK-2400,
Denmark.
| | - Peter Schjerling
- Institute of Sports Medicine Copenhagen,
Department of Orthopedic Surgery M81, Bispebjerg and Frederiksberg Hospital,
Copenhagen, Denmark,Center for Healthy Aging, Faculty of
Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael R. Krogsgaard
- Section for Sports Traumatology M51,
Department of Orthopedic Surgery, Bispebjerg and Frederiksberg Hospital, Copenhagen,
Denmark
| | - Michael M. Petersen
- Musculoskeletal Tumor Section,
Department of Orthopedic Surgery, Rigshospitalet, Faculty of Health and Medical
Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Olsen
- Aarhus AMS Centre (AARAMS), Department
of Physics and Astronomy, Aarhus University, Aarhus C, Denmark
| | - Michael Kjær
- Institute of Sports Medicine Copenhagen,
Department of Orthopedic Surgery M81, Bispebjerg and Frederiksberg Hospital,
Copenhagen, Denmark,Center for Healthy Aging, Faculty of
Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katja M. Heinemeier
- Institute of Sports Medicine Copenhagen,
Department of Orthopedic Surgery M81, Bispebjerg and Frederiksberg Hospital,
Copenhagen, Denmark,Center for Healthy Aging, Faculty of
Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
46
|
Revell CK, Jensen OE, Shearer T, Lu Y, Holmes DF, Kadler KE. Collagen fibril assembly: New approaches to unanswered questions. Matrix Biol Plus 2021; 12:100079. [PMID: 34381990 PMCID: PMC8334717 DOI: 10.1016/j.mbplus.2021.100079] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/22/2022] Open
Abstract
Collagen fibrils are essential for metazoan life. They are the largest, most abundant, and most versatile protein polymers in animals, where they occur in the extracellular matrix to form the structural basis of tissues and organs. Collagen fibrils were first observed at the turn of the 20th century. During the last 40 years, the genes that encode the family of collagens have been identified, the structure of the collagen triple helix has been solved, the many enzymes involved in the post-translational modifications of collagens have been identified, mutations in the genes encoding collagen and collagen-associated proteins have been linked to heritable disorders, and changes in collagen levels have been associated with a wide range of diseases, including cancer. Yet despite extensive research, a full understanding of how cells assemble collagen fibrils remains elusive. Here, we review current models of collagen fibril self-assembly, and how cells might exert control over the self-assembly process to define the number, length and organisation of fibrils in tissues.
Collapse
Affiliation(s)
- Christopher K. Revell
- Department of Mathematics, University of Manchester, Alan Turing Building, Oxford Road, Manchester M13 9PL, UK
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Oliver E. Jensen
- Department of Mathematics, University of Manchester, Alan Turing Building, Oxford Road, Manchester M13 9PL, UK
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Tom Shearer
- Department of Mathematics, University of Manchester, Alan Turing Building, Oxford Road, Manchester M13 9PL, UK
- Department of Materials, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Yinhui Lu
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - David F. Holmes
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Karl E. Kadler
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
47
|
Whittaker JL, Runhaar J, Bierma-Zeinstra S, Roos EM. A lifespan approach to osteoarthritis prevention. Osteoarthritis Cartilage 2021; 29:1638-1653. [PMID: 34560260 DOI: 10.1016/j.joca.2021.06.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/27/2021] [Accepted: 06/18/2021] [Indexed: 02/02/2023]
Abstract
Prevention is an attractive solution for the staggering and increasingly unmanageable burden of osteoarthritis. Despite this, the field of osteoarthritis prevention is relatively immature. To date, most of what is known about preventing osteoarthritis and risk factors for osteoarthritis is relative to the disease (underlying biology and pathophysiology) of osteoarthritis, with few studies considering risk factors for osteoarthritis illness, the force driving the personal, financial and societal burden. In this narrative review we will discuss what is known about osteoarthritis prevention, propose actionable prevention strategies related to obesity and joint injury which have emerged as important modifiable risk factors, identify where evidence is lacking, and give insight into what might be possible in terms of prevention by focussing on a lifespan approach to the illness of osteoarthritis, as opposed to a structural disease of the elderly. By targeting a non-specialist audience including scientists, clinicians, students, industry employees and others that are interested in osteoarthritis but who do not necessarily focus on osteoarthritis, the goal is to generate discourse and motivate inquiry which propel the field of osteoarthritis prevention into the mainstream.
Collapse
Affiliation(s)
- J L Whittaker
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Arthritis Research Canada, Canada.
| | - J Runhaar
- Erasmus MC University Medical Center Rotterdam, Department of General Practice, Rotterdam, the Netherlands.
| | - S Bierma-Zeinstra
- Erasmus MC University Medical Center Rotterdam, Department of General Practice, Rotterdam, the Netherlands; Erasmus MC University Medical Center Rotterdam, Department of General Practice, and Department of Orthopaedics, Rotterdam, the Netherlands.
| | - E M Roos
- Department of Sports Science and Clinical Biomechanics, Musculoskeletal Function and Physiotherapy, University of Southern Denmark, Denmark.
| |
Collapse
|
48
|
Paatela T, Vasara A, Sormaala M, Nurmi H, Kautiainen H, Kiviranta I. Chondral and Osteochondritis Dissecans Lesions Treated by Autologous Chondrocytes Implantation: A Mid- to Long-Term Nonrandomized Comparison. Cartilage 2021; 13:1105S-1112S. [PMID: 32602351 PMCID: PMC8808789 DOI: 10.1177/1947603520935953] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE The aim of this study was to compare the clinical outcome of cartilage repair with autologous chondrocyte implantation (ACI) in patients with osteochondritis dissecans (OCD) lesions and full-thickness cartilage lesions. DESIGN This study included a cohort of 115 consecutive patients with a cartilage lesion of the knee treated with ACI. Of the patients, 35 had an OCD lesion and 80 a full-thickness cartilage lesion. During a follow-up period from 2 to 13 years all treatment failures were identified. The failure rate between OCD lesions and full-thickness cartilage lesions was compared with Kaplan-Meier analysis. Patient-reported outcome was evaluated 2 years postoperatively with the Lysholm score. RESULTS During the follow-up 21 out of 115 patients encountered a treatment failure. The failure rate for full-thickness cartilage lesions was 19.1% and for OCD lesions 43.3% over the 10-year follow-up. Patient-reported outcome improved from baseline to 2 years postoperatively. The improvement from baseline was statistically significant, and the Lysholm score improved more than the minimal clinically important difference. The patient-reported outcome showed no difference between lesion types at 2 years. CONCLUSIONS In the presented retrospective study, the failure rate of first-generation ACI was higher in OCD lesions than in large full-thickness cartilage lesions, suggesting that OCD lesions may associate with properties that affect the durability of repair tissue. Future prospective studies are needed to tell us how to best repair OCD lesions with biological tissue engineering.
Collapse
Affiliation(s)
- Teemu Paatela
- Department of Orthopaedics and
Traumatology, Helsinki University Hospital, Helsinki, Finland,Teemu Paatela, Department of Orthopaedics
and Traumatology, Helsinki University Hospital, PO Box 900, Topeliuksenkatu 5,
Helsinki FI-00029 HUS, Finland.
| | - Anna Vasara
- Department of Orthopaedics and
Traumatology, Helsinki University Hospital, Helsinki, Finland
| | - Markus Sormaala
- Department of Radiology, Helsinki
University Hospital, Helsinki, Finland
| | - Heikki Nurmi
- Department of Orthopaedics and
Traumatology, Central Finland Central Hospital, Jyväskylä, Finland
| | - Hannu Kautiainen
- Primary Health Care Unit, Kuopio
University Hospital, Kuopio, Finland,Folkhälsan Research Center, Helsinki,
Finland
| | - Ilkka Kiviranta
- Department of Orthopaedics and
Traumatology, Helsinki University Hospital, Helsinki, Finland,Department of Orthopaedics and
Traumatology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
49
|
Styczynska-Soczka K, Amin AK, Simpson AHW, Hall AC. Optimization and Validation of a Human Ex Vivo Femoral Head Model for Preclinical Cartilage Research and Regenerative Therapies. Cartilage 2021; 13:386S-397S. [PMID: 32567330 PMCID: PMC8721618 DOI: 10.1177/1947603520934534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Articular cartilage is incapable of effective repair following injury or during osteoarthritis. While there have been developments in cartilage repair technologies, there is a need to advance biologically relevant models for preclinical testing of biomaterial and regenerative therapies. This study describes conditions for the effective ex vivo culture of the whole human femoral head. DESIGN Fresh, viable femoral heads were obtained from femoral neck fractures and cultured for up to 10 weeks in (a) Dulbecco's modified Eagle's medium (DMEM); (b) DMEM + mixing; (c) DMEM + 10% human serum (HS); (d) DMEM + 10% HS + mixing. The viability, morphology, volume, and density of fluorescently labelled in situ chondrocytes and cartilage surface roughness were assessed by confocal microscopy. Cartilage histology was studied for glycosaminoglycan content using Alcian blue and collagen content using picrosirius red. RESULTS Chondrocyte viability remained at >95% in DMEM + 10% HS. In DMEM alone, viability remained high for ~4 weeks and then declined. For the other conditions, superficial zone chondrocyte viability fell to <35% at 10 weeks with deeper zones being relatively unaffected. In DMEM + 10% HS at 10 weeks, the number of chondrocytes possessing cytoplasmic processes increased compared with DMEM (P = 0.017). Alcian blue labeling decreased (P = 0.02) and cartilage thinned (P ≤ 0.05); however, there was no change to surface roughness, chondrocyte density, chondrocyte volume, or picrosirius red labeling (P > 0.05). CONCLUSIONS In this ex vivo model, chondrocyte viability was maintained in human femoral heads for up to 10 weeks in culture, a novel finding not previously reported. This human model could prove invaluable for the exploration, development, and assessment of preclinical cartilage repair and regenerative therapies.
Collapse
Affiliation(s)
| | - Anish K. Amin
- Department of Trauma and Orthopaedic
Surgery, Royal Infirmary of Edinburgh, Edinburgh, Scotland, UK
| | - A. Hamish W. Simpson
- Department of Trauma and Orthopaedic
Surgery, Royal Infirmary of Edinburgh, Edinburgh, Scotland, UK
| | - Andrew C. Hall
- Biomedical Sciences, Edinburgh Medical
School, University of Edinburgh, Edinburgh, Scotland, UK,Andrew C. Hall, Biomedical Sciences,
Edinburgh Medical School, University of Edinburgh, Hugh Robson Building, George
Square, Edinburgh, EH8 9XD, Scotland, UK.
| |
Collapse
|
50
|
Rolfs Z, Frey BL, Shi X, Kawai Y, Smith LM, Welham NV. An atlas of protein turnover rates in mouse tissues. Nat Commun 2021; 12:6778. [PMID: 34836951 PMCID: PMC8626426 DOI: 10.1038/s41467-021-26842-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/25/2021] [Indexed: 01/25/2023] Open
Abstract
Protein turnover is critical to cellular physiology as well as to the growth and maintenance of tissues. The unique synthesis and degradation rates of each protein help to define tissue phenotype, and knowledge of tissue- and protein-specific half-lives is directly relevant to protein-related drug development as well as the administration of medical therapies. Using stable isotope labeling and mass spectrometry, we determine the in vivo turnover rates of thousands of proteins-including those of the extracellular matrix-in a set of biologically important mouse tissues. We additionally develop a data visualization platform, named ApplE Turnover, that enables facile searching for any protein of interest in a tissue of interest and then displays its half-life, confidence interval, and supporting measurements. This extensive dataset and the corresponding visualization software provide a reference to guide future studies of mammalian protein turnover in response to physiologic perturbation, disease, or therapeutic intervention.
Collapse
Affiliation(s)
- Zach Rolfs
- grid.14003.360000 0001 2167 3675Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Brian L. Frey
- grid.14003.360000 0001 2167 3675Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Xudong Shi
- grid.14003.360000 0001 2167 3675Division of Otolaryngology–Head and Neck Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
| | - Yoshitaka Kawai
- grid.14003.360000 0001 2167 3675Division of Otolaryngology–Head and Neck Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA ,grid.258799.80000 0004 0372 2033Present Address: Department of Otolaryngology–Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507 Japan
| | - Lloyd M. Smith
- grid.14003.360000 0001 2167 3675Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Nathan V. Welham
- grid.14003.360000 0001 2167 3675Division of Otolaryngology–Head and Neck Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
| |
Collapse
|