1
|
Holiuk Y, Birsa R, Bukreieva T, Nemtinov P, Kyryk V, Ustymenko A, Mazevych V, Sokolov M, Lobyntseva G, Shablii V. Effectiveness and safety of multiple injections of human placenta-derived MSCs for knee osteoarthritis: a nonrandomized phase I trial. BMC Musculoskelet Disord 2025; 26:418. [PMID: 40281581 PMCID: PMC12032682 DOI: 10.1186/s12891-025-08664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
OBJECTIVE This study investigates the safety and efficacy of three intra-articular (IA) injections of cryopreserved human placenta-derived mesenchymal stem cells (hP-MSCs) for knee osteoarthritis (KOA) over a 1-year follow-up period. METHODS A total of 26 patients with stage II-III KOA were enrolled in this non-randomized, open-label study. Patients received either conventional therapy with hyaluronic acid (HA) alone (Control group, n = 11) or in combination with hP-MSCs (MSC group, n = 15) via three intra-articular injections with 4-week intervals. Clinical outcomes were assessed using the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), Visual Analogue Scale (VAS), and magnetic resonance imaging (MRI) at 6 and 12 months following the first injection. Blood samples were analyzed for cytokine levels. RESULTS Three injections of hP-MSCs combined with HA were well-tolerated, with no severe adverse events observed. Significant improvements in WOMAC and VAS scores were noted in the MSC group compared to the Control group at both 6 and 12 months. MRI analysis revealed no significant differences in cartilage thickness or optical density index between the groups. Additionally, serum cytokine analysis showed a significant decrease in interleukin-2 (IL-2) levels in the MSC group, indicating an anti-inflammatory effect of hP-MSCs. However, no significant changes were observed in other cytokines. CONCLUSION This study demonstrates that three intra-articular injections of cryopreserved hP-MSCs in combination with HA are safe and effective for treating KOA, providing sustained clinical improvement at the 1-year follow-up. TRIAL REGISTRATION NCT04453111, #7/09.26.2018. Registered 02 January 2020, https://www. CLINICALTRIALS gov/study/NCT04453111 .
Collapse
Affiliation(s)
- Yevhen Holiuk
- State Institution "The Institute of Traumatology and Orthopedics by NAMS of Ukraine", 27 Bulvarno-Kudriavska Street, Kyiv, 01601, Ukraine
| | - Roman Birsa
- Department of Traumatology, Kyiv City Clinical Hospital, #6, 3 Guzara Ave, Kyiv, 03680, Ukraine
| | - Tetiana Bukreieva
- Laboratory of Biosynthesis of Nucleic Acids, Institute of Molecular Biology and Genetics of National Academy of Science of Ukraine, 150 Zabolotnogo Str, Kyiv, 03143, Ukraine
- Placenta Stem Cell Laboratory, Institute of Cell Therapy, 9 Mokra str, Cryobank, Kyiv, 03035, Ukraine
| | - Petro Nemtinov
- Institute of Cell Therapy, 9 Mokra str, Kyiv, 03035, Ukraine
| | - Vitalii Kyryk
- Cell and Tissue Technologies Department, M. D. Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine of the National Academy of Medical Sciences of Ukraine, 5 Svyatoslav Khorobrygo str, Kyiv, 03151, Ukraine
- Laboratory of Pathological Physiology and Immunology, D. F. Chebotarev Institute of Gerontology of the National Academy of Medical Sciences of Ukraine, 67 Vyshgorodska Street, Kyiv, 04114, Ukraine
| | - Alina Ustymenko
- Cell and Tissue Technologies Department, M. D. Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine of the National Academy of Medical Sciences of Ukraine, 5 Svyatoslav Khorobrygo str, Kyiv, 03151, Ukraine
- Laboratory of Pathological Physiology and Immunology, D. F. Chebotarev Institute of Gerontology of the National Academy of Medical Sciences of Ukraine, 67 Vyshgorodska Street, Kyiv, 04114, Ukraine
| | - Vadym Mazevych
- State Institution "The Institute of Traumatology and Orthopedics by NAMS of Ukraine", 27 Bulvarno-Kudriavska Street, Kyiv, 01601, Ukraine
| | - Mykola Sokolov
- Institute of Cell Therapy, 9 Mokra str, Kyiv, 03035, Ukraine
| | | | - Volodymyr Shablii
- Placenta Stem Cell Laboratory, Institute of Cell Therapy, 9 Mokra str, Cryobank, Kyiv, 03035, Ukraine.
- Department of Protein Synthesis Enzymology, Institute of Molecular Biology and Genetics of National Academy of Science of Ukraine, 150 Zabolotnogo Str, 03143, Kyiv, Ukraine.
| |
Collapse
|
2
|
Takase K, McCulloch PC, Yik JHN, Haudenschild DR. Clinical and molecular landscape of post-traumatic osteoarthritis. Connect Tissue Res 2025:1-7. [PMID: 40265194 DOI: 10.1080/03008207.2025.2490797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage breakdown, chronic pain, and disability. Post-traumatic osteoarthritis (PTOA), a secondary form of OA, arises from joint injuries and consistently accounts for a proportion of symptomatic cases. Unlike primary OA, PTOA has a well-defined initiation point, presenting an opportunity for early intervention. Over the past two decades, research has shifted from a cartilage-centric view to a broader understanding of OA as a multifaceted disease involving inflammation, oxidative stress, and complex molecular crosstalk between chondrocytes, synoviocytes, osteocytes, and immune cells. Key inflammatory mediators, such as IL-1β, IL-6, TNF-α, and Wnt/β-catenin signaling, drive disease progression. Advances in imaging, biomarker discovery, and animal models have provided insights into early disease mechanisms. However, gaps remain in understanding the molecular events that trigger PTOA onset, the interplay between joint tissues, and the identification of reliable early biomarkers. Delayed diagnosis, lack of disease-modifying therapies, and OA's complexity remain critical barriers. Future directions should focus on precision medicine integrating biomarkers, imaging, and artificial intelligence for early diagnosis and risk stratification. Emerging regenerative and gene therapies, while promising, would benefit from moving beyond single-pathway targeting, as OA's multifaceted nature makes a combination approach desirable to simultaneously address inflammation, oxidative stress, cartilage matrix degradation, and tissue repair. Multidisciplinary collaborations between clinicians, molecular biologists, and bioengineers are essential to translating discoveries into effective interventions. A paradigm shift toward early, personalized treatment strategies is necessary to improve long-term outcomes in PTOA and OA management.
Collapse
Affiliation(s)
- Kyohei Takase
- Department of Translational Orthopedic Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Patrick C McCulloch
- Houston Methodist Orthopedics & Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Jasper H N Yik
- Department of Translational Orthopedic Research, Houston Methodist Research Institute, Houston, TX, USA
| | - Dominik R Haudenschild
- Department of Translational Orthopedic Research, Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Orthopedics & Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
3
|
Deng M, Tang C, Yin L, Jiang Y, Huang Y, Feng Y, Chen C. Clinical and omics biomarkers in osteoarthritis diagnosis and treatment. J Orthop Translat 2025; 50:295-305. [PMID: 39911590 PMCID: PMC11795539 DOI: 10.1016/j.jot.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/03/2024] [Accepted: 12/09/2024] [Indexed: 02/07/2025] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease that significantly impacts the quality of life for hundreds of millions, and is a major cause of disability. Despite this, diagnostic and therapeutic options for OA are still limited. With advances in molecular biology, an increasing number of OA biomarkers have been identified, which not only enhances our understanding of OA pathogenesis, but also offers new approaches for OA diagnosis and treatment. This review discussed the research progress on traditional OA biomarkers, and analyzed the application of various omics, including genomics, transcriptomics, proteomics, and metabolomics, in the diagnosis and treatment of OA. Furthermore, we explored how integrating multi-omics methods can reveal interactions among different biomolecules and their roles in the development of OA. This emerging interdisciplinary approach not only provides a more comprehensive understanding of the fundamental biological characteristics of OA, but also aids in identifying new integrated biomarkers, thereby allowing for more accurate predictions of disease progression and treatment responses. The identification and development of biomarkers offer new perspectives in understanding OA, enhancing the specificity and sensitivity of biological diagnostic markers, providing a basis for the design of targeted drugs, and ultimately advancing the development of precision diagnosis and treatment strategies in clinical OA. This study provides an overview of both commonly used and emerging biomarkers of OA which is beneficial for a more accurate, timely, effective clinical diagnosis and treatment for OA.
Collapse
Affiliation(s)
- Muhai Deng
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Cong Tang
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Li Yin
- Department of Orthopaedics, General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Yunsheng Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yang Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yong Feng
- Department of Orthopedic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Cheng Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
4
|
Hutcherson C, Luke B, Khader K, Dhaher YY. Unraveling the complex interplay of sex, endocrinology, and inflammation in post-Injury articular cartilage breakdown through in silico modeling. Sci Rep 2024; 14:28654. [PMID: 39562596 PMCID: PMC11576913 DOI: 10.1038/s41598-024-77730-x] [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: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/21/2024] Open
Abstract
The onset of degenerative joint diseases such as post-traumatic osteoarthritis (PTOA) are associated with joint injury, biomechanical changes, and synovial biochemical anomalies. Sex and reproductive endocrinology have been emerging as potential risk factors, with epidemiological evidence revealing that female's exhibit higher PTOA risk and poorer outcomes post-injury compared to males. Sex hormones, including estradiol, progesterone, and testosterone, have been shown to regulate inflammatory signaling in immune and synovial cells, yet their collective impact on injury-induced joint inflammation and catabolism is poorly understood. Using an in silico kinetic model, we investigated the effects of sex-specific endocrine states on post-injury mechanisms in the human synovial joint. Our model results reveal that heightened estradiol levels in pre-menopausal females during the peri-ovulatory phase increase interleukin (IL)-1β expression and suppress IL-10 expression within the synovium after a simulated injury. Conversely, elevated testosterone levels in males decrease post-injury IL-1β, tumor necrosis factor alpha (TNF)-α, and stromelysin (MMP)-3 expression while increasing IL-10 production compared to females. Gaining insight into the effects of sex hormones on injury-induced inflammation and cartilage degradation provides a basis for designing future experimental and clinical studies to explore their effects on the synovial system, with a particular focus on the female sex.
Collapse
Affiliation(s)
- C Hutcherson
- Department of Physical Medicine and Rehabilitation, University of Texas Southwestern, Dallas, TX, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - B Luke
- Department of Mechanical Engineering, Valparaiso University, Valparaiso, IN, USA
| | - K Khader
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Y Y Dhaher
- Department of Physical Medicine and Rehabilitation, University of Texas Southwestern, Dallas, TX, USA.
- Department of Orthopaedic Surgery, University of Texas Southwestern, Dallas, TX, USA.
| |
Collapse
|
5
|
Deng Y, Perry TA, Hulley P, Maciewicz RA, Mitchelmore J, Perry D, Larsson S, Brachat S, Struglics A, Appleton CT, Kluzek S, Arden NK, Felson D, Marsden B, Tom BDM, Bondi L, Kapoor M, Batchelor V, Mackay-Alderson J, Kumar V, Lohmander LS, Welting TJ, Walsh DA, Valdes AM, Vincent TL, Watt FE, Jostins-Dean L. Development of methodology to support molecular endotype discovery from synovial fluid of individuals with knee osteoarthritis: The STEpUP OA consortium. PLoS One 2024; 19:e0309677. [PMID: 39556578 PMCID: PMC11573211 DOI: 10.1371/journal.pone.0309677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 08/15/2024] [Indexed: 11/20/2024] Open
Abstract
OBJECTIVES To develop a protocol for largescale analysis of synovial fluid proteins, for the identification of biological networks associated with subtypes of osteoarthritis. METHODS Synovial Fluid To detect molecular Endotypes by Unbiased Proteomics in Osteoarthritis (STEpUP OA) is an international consortium utilising clinical data (capturing pain, radiographic severity and demographic features) and knee synovial fluid from 17 participating cohorts. 1746 samples from 1650 individuals comprising OA, joint injury, healthy and inflammatory arthritis controls, divided into discovery (n = 1045) and replication (n = 701) datasets, were analysed by SomaScan Discovery Plex V4.1 (>7000 SOMAmers/proteins). An optimised approach to standardisation was developed. Technical confounders and batch-effects were identified and adjusted for. Poorly performing SOMAmers and samples were excluded. Variance in the data was determined by principal component (PC) analysis. RESULTS A synovial fluid standardised protocol was optimised that had good reliability (<20% co-efficient of variation for >80% of SOMAmers in pooled samples) and overall good correlation with immunoassay. 1720 samples and >6290 SOMAmers met inclusion criteria. 48% of data variance (PC1) was strongly correlated with individual SOMAmer signal intensities, particularly with low abundance proteins (median correlation coefficient 0.70), and was enriched for nuclear and non-secreted proteins. We concluded that this component was predominantly intracellular proteins, and could be adjusted for using an 'intracellular protein score' (IPS). PC2 (7% variance) was attributable to processing batch and was batch-corrected by ComBat. Lesser effects were attributed to other technical confounders. Data visualisation revealed clustering of injury and OA cases in overlapping but distinguishable areas of high-dimensional proteomic space. CONCLUSIONS We have developed a robust method for analysing synovial fluid protein, creating a molecular and clinical dataset of unprecedented scale to explore potential patient subtypes and the molecular pathogenesis of OA. Such methodology underpins the development of new approaches to tackle this disease which remains a huge societal challenge.
Collapse
Affiliation(s)
- Yun Deng
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Thomas A. Perry
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Philippa Hulley
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Rose A. Maciewicz
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | | | - Darryl Perry
- SomaLogic, Boulder, Colorado, United States of America
| | - Staffan Larsson
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sophie Brachat
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - André Struglics
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - C. Thomas Appleton
- Bone and Joint Institute, University of Western Ontario, London, Ontario, Canada
| | - Stefan Kluzek
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
- NIHR Nottingham Biomedical Research Centre and Versus Arthritis Sport, Exercise and Osteoarthritis Centre, University of Nottingham, Nottingham, United Kingdom
| | - Nigel K. Arden
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
- Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, University of Oxford, Oxford, United Kingdom
| | - David Felson
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Brian Marsden
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Brian D. M. Tom
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Laura Bondi
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Mohit Kapoor
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada
| | - Vicky Batchelor
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Jennifer Mackay-Alderson
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Vinod Kumar
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - L. Stefan Lohmander
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Tim J. Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, Netherlands
| | - David A. Walsh
- Pain Centre Versus Arthritis, Advanced Pain Discovery Platform, and the NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom
- Sherwood Forest Hospitals NHS Foundation Trust, Sutton in Ashfield, United Kingdom
| | - Ana M. Valdes
- Pain Centre Versus Arthritis, Advanced Pain Discovery Platform, and the NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom
| | | | - Tonia L. Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Fiona E. Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Luke Jostins-Dean
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
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
|
7
|
Gilbert SJ, Soul J, Hao Y, Lin H, Piróg KA, Coxhead J, Patel K, Barter MJ, Young DA, Blain EJ. Comparative transcriptomic analysis of articular cartilage of post-traumatic osteoarthritis models. Dis Model Mech 2024; 17:dmm050583. [PMID: 39314058 PMCID: PMC11524441 DOI: 10.1242/dmm.050583] [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: 11/06/2023] [Accepted: 09/17/2024] [Indexed: 09/25/2024] Open
Abstract
Animal models of post-traumatic osteoarthritis (PTOA) recapitulate the pathological changes observed in human PTOA. Here, skeletally mature C57Bl6 mice were subjected to either rapid-onset non-surgical mechanical rupture of the anterior cruciate ligament (ACL) or to surgical destabilisation of the medial meniscus (DMM). Transcriptome profiling of micro-dissected cartilage at day 7 or day 42 following ACL or DMM procedure, respectively, showed that the two models were comparable and highly correlative. Gene ontology (GO) enrichment analysis identified similarly enriched pathways that were overrepresented by anabolic terms. To address the transcriptome changes more completely in the ACL model, we also performed small RNA sequencing, describing the first microRNA profile of this model. miR-199-5p was amongst the most abundant, yet differentially expressed, microRNAs, and its inhibition in primary human chondrocytes led to a transcriptome response that was comparable to that observed in both human 'OA damaged vs intact cartilage' and murine DMM cartilage datasets. We also experimentally verified CELSR1, GIT1, ECE1 and SOS2 as novel miR-199-5p targets. Together, these data support the use of the ACL rupture model as a non-invasive companion to the DMM model.
Collapse
Affiliation(s)
- Sophie J. Gilbert
- Biomechanics and Bioengineering Research Centre Versus Arthritis, Biomedicine Division, School of Biosciences, The Sir Martin Evans Building, Cardiff University, Cardiff CF10 3AX, Wales, UK
| | - Jamie Soul
- Biosciences Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon TyneNE1 3BZ, UK
| | - Yao Hao
- Biosciences Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon TyneNE1 3BZ, UK
| | - Hua Lin
- Biosciences Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon TyneNE1 3BZ, UK
| | - Katarzyna A. Piróg
- Biosciences Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon TyneNE1 3BZ, UK
| | - Jonathan Coxhead
- Biosciences Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon TyneNE1 3BZ, UK
| | - Krutik Patel
- Biosciences Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon TyneNE1 3BZ, UK
| | - Matt J. Barter
- Biosciences Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon TyneNE1 3BZ, UK
| | - David A. Young
- Biosciences Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon TyneNE1 3BZ, UK
| | - Emma J. Blain
- Biomechanics and Bioengineering Research Centre Versus Arthritis, Biomedicine Division, School of Biosciences, The Sir Martin Evans Building, Cardiff University, Cardiff CF10 3AX, Wales, UK
| |
Collapse
|
8
|
Ayala S, Matan SO, Delco ML, Fortier LA, Cohen I, Bonassar LJ. Degradation of lubricating molecules in synovial fluid alters chondrocyte sensitivity to shear strain. J Orthop Res 2024. [PMID: 39182184 DOI: 10.1002/jor.25960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/01/2024] [Accepted: 08/03/2024] [Indexed: 08/27/2024]
Abstract
Articular joints facilitate motion and transfer loads to underlying bone through a combination of cartilage tissue and synovial fluid, which together generate a low-friction contact surface. Traumatic injury delivered to cartilage and the surrounding joint capsule causes secretion of proinflammatory cytokines by chondrocytes and the synovium, triggering cartilage matrix breakdown and impairing the ability of synovial fluid to lubricate the joint. Once these inflammatory processes become chronic, posttraumatic osteoarthritis (PTOA) development begins. However, the exact mechanism by which negative alterations to synovial fluid leads to PTOA pathogenesis is not fully understood. We hypothesize that removing the lubricating macromolecules from synovial fluid alters the relationship between mechanical loads and subsequent chondrocyte behavior in injured cartilage. To test this hypothesis, we utilized an ex vivo model of PTOA that involves subjecting cartilage explants to a single rapid impact followed by continuous articulation within a lubricating bath of either healthy synovial fluid, phosphate-buffered saline (PBS), synovial fluid treated with hyaluronidase, or synovial fluid treated with trypsin. These treatments degrade the main macromolecules attributed with providing synovial fluid with its lubricating properties; hyaluronic acid and lubricin. Explants were then bisected and fluorescently stained to assess global and depth-dependent cell death, caspase activity, and mitochondrial depolarization. Explants were tested via confocal elastography to determine the local shear strain profile generated in each lubricant. These results show that degrading hyaluronic acid or lubricin in synovial fluid significantly increases middle zone chondrocyte damage and shear strain loading magnitudes, while also altering chondrocyte sensitivity to loading.
Collapse
Affiliation(s)
- Steven Ayala
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Salman O Matan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Michelle L Delco
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Lisa A Fortier
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Itai Cohen
- Department of Physics, Cornell University, Ithaca, New York, USA
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
9
|
Švajger U, Kolar M, Kobold A, Drobnič M. Mechanisms of treatment effects using allogeneic, umbilical cord-derived mesenchymal stromal stem cells (MSCs) in knee osteoarthritis: a pharmacological clinical study protocol. Trials 2024; 25:533. [PMID: 39135209 PMCID: PMC11320946 DOI: 10.1186/s13063-024-08360-x] [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: 03/15/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) presents a prevalent orthopedic condition causing substantial impairment in the quality of life and imposing a significant societal and economic burden. Mesenchymal stromal/stem cells (MSCs), known for their regenerative properties and immunomodulatory effects, have emerged as a promising therapeutic avenue in regenerative medicine. Despite MSCs' therapeutic potential, their precise mechanisms of action in KOA remain underexplored. METHODS Conducted as a randomized, open-label clinical trial, 20 patients will be enrolled, with 10 in the intervention group and 10 in the control group. The primary focus will be to explore the molecular mechanisms associated with MSC therapy. Biomarkers and gene expressions related to cartilage metabolism, inflammation, immune modulation, and pain in the synovial fluid, blood, and tissue samples will be analyzed. Patients will undergo pre- and post-treatment evaluations using patient-reported outcome measures (PROMs) and comprehensive clinical assessments. DISCUSSION This is an exploratory study with the goal to provide comprehensive insights into the therapeutic effects of MSCs on a molecular level, potentially paving the way for optimized and more effective MSC-based therapies in the management of KOA, as well as furthering the development of novel treatment strategies. TRIAL REGISTRATION ClinicalTrials.gov, NCT06078059. Registered on 5 October 2023.
Collapse
Affiliation(s)
- Urban Švajger
- Slovenian Institute for Transfusion Medicine, Šlajmerjeva cesta 6, Ljubljana, 1000, Slovenia.
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana, 1000, Slovenia.
| | - Matic Kolar
- Department of Orthopaedic Surgery, University Medical Centre Ljubljana, Zaloška cesta 9, Ljubljana, 1000, Slovenia
- Chair of Orthopaedics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, 1000, Slovenia
| | - Andrej Kobold
- Slovenian Institute for Transfusion Medicine, Šlajmerjeva cesta 6, Ljubljana, 1000, Slovenia
| | - Matej Drobnič
- Department of Orthopaedic Surgery, University Medical Centre Ljubljana, Zaloška cesta 9, Ljubljana, 1000, Slovenia
- Chair of Orthopaedics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, 1000, Slovenia
| |
Collapse
|
10
|
Gilbert SJ, Jones R, Egan BJ, Bonnet CS, Evans SL, Mason DJ. Investigating mechanical and inflammatory pathological mechanisms in osteoarthritis using MSC-derived osteocyte-like cells in 3D. Front Endocrinol (Lausanne) 2024; 15:1359052. [PMID: 39157681 PMCID: PMC11328832 DOI: 10.3389/fendo.2024.1359052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/17/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Changes to bone physiology play a central role in the development of osteoarthritis with the mechanosensing osteocyte releasing factors that drive disease progression. This study developed a humanised in vitro model to detect osteocyte responses to either interleukin-6, a driver of degeneration and bone remodelling in animal and human joint injury, or mechanical loading, to mimic osteoarthritis stimuli in joints. Methods Human MSC cells (Y201) were differentiated in 3-dimensional type I collagen gels in osteogenic media and osteocyte phenotype assessed by RTqPCR and immunostaining. Gels were subjected to a single pathophysiological load or stimulated with interleukin-6 with unloaded or unstimulated cells as controls. RNA was extracted 1-hour post-load and assessed by RNAseq. Markers of pain, bone remodelling, and inflammation were quantified by RT-qPCR and ELISA. Results Y201 cells embedded within 3D collagen gels assumed dendritic morphology and expressed mature osteocytes markers. Mechanical loading of the osteocyte model regulated 7564 genes (Padj p<0.05, 3026 down, 4538 up). 93% of the osteocyte transcriptome signature was expressed in the model with 38% of these genes mechanically regulated. Mechanically loaded osteocytes regulated 26% of gene ontology pathways linked to OA pain, 40% reflecting bone remodelling and 27% representing inflammation. Load regulated genes associated with osteopetrosis, osteoporosis and osteoarthritis. 42% of effector genes in a genome-wide association study meta-analysis were mechanically regulated by osteocytes with 10 genes representing potential druggable targets. Interleukin-6 stimulation of osteocytes at concentrations reported in human synovial fluids from patients with OA or following knee injury, regulated similar readouts to mechanical loading including markers of pain, bone remodelling, and inflammation. Discussion We have developed a reproducible model of human osteocyte like cells that express >90% of the genes in the osteocyte transcriptome signature. Mechanical loading and inflammatory stimulation regulated genes and proteins implicated in osteoarthritis symptoms of pain as well as inflammation and degeneration underlying disease progression. Nearly half of the genes classified as 'effectors' in GWAS were mechanically regulated in this model. This model will be useful in identifying new mechanisms underlying bone and joint pathologies and testing drugs targeting those mechanisms.
Collapse
Affiliation(s)
- Sophie J. Gilbert
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Ryan Jones
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Ben J. Egan
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Cleo Selina Bonnet
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Sam L. Evans
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Engineering, Cardiff University, Cardiff, United Kingdom
| | - Deborah J. Mason
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
11
|
Kraus VB, Hsueh MF. Molecular biomarker approaches to prevention of post-traumatic osteoarthritis. Nat Rev Rheumatol 2024; 20:272-289. [PMID: 38605249 DOI: 10.1038/s41584-024-01102-y] [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: 03/05/2024] [Indexed: 04/13/2024]
Abstract
Up to 50% of individuals develop post-traumatic osteoarthritis (PTOA) within 10 years following knee-joint injuries such as anterior cruciate ligament rupture or acute meniscal tear. Lower-extremity PTOA prevalence is estimated to account for ≥12% of all symptomatic osteoarthritis (OA), or approximately 5.6 million cases in the USA. With knowledge of the inciting event, it might be possible to 'catch PTOA in the act' with sensitive imaging and soluble biomarkers and thereby prevent OA sequelae by early intervention. Existing biomarker data in the joint-injury literature can provide insights into the pathogenesis and early risk trajectory related to PTOA and can help to elucidate a research agenda for preventing or slowing the onset of PTOA. Non-traumatic OA and PTOA have many clinical, radiological and genetic similarities, and efforts to understand early risk trajectories in PTOA might therefore contribute to the identification and classification of early non-traumatic OA, which is the most prevalent form of OA.
Collapse
Affiliation(s)
- Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA.
| | - Ming-Feng Hsueh
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
12
|
Turnbull J, Jha RR, Barrett DA, Valdes AM, Alderson J, Williams A, Vincent TL, Watt FE, Chapman V. The Effect of Acute Knee Injuries and Related Knee Surgery on Serum Levels of Pro- and Anti-inflammatory Lipid Mediators and Their Associations With Knee Symptoms. Am J Sports Med 2024; 52:987-997. [PMID: 38406872 PMCID: PMC10943603 DOI: 10.1177/03635465241228209] [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: 02/16/2023] [Accepted: 11/29/2023] [Indexed: 02/27/2024]
Abstract
BACKGROUND Despite an acute knee injury being a major risk factor for osteoarthritis, the factors that initiate and maintain this risk of longer-term knee symptoms are poorly understood. Bioactive lipids derived from omega-3 and -6 polyunsaturated fatty acids have key roles in the regulation of the inflammatory response and have been linked to joint damage and osteoarthritis pain in translational models. HYPOTHESIS There would be associations between systemic levels of bioactive lipids and knee symptoms longitudinally after an acute knee injury and related knee surgery. STUDY DESIGN Controlled laboratory study. METHODS This study analyzed a subset of young, active adults who had sustained an acute knee injury (recruited via a surgical care pathway) and healthy age- and sex-matched controls. Surgery, if performed, was conducted after the baseline serum sample was taken and before the 3-month and 2-year visits. Liquid chromatography-tandem mass spectrometry of 41 bioactive lipids was carried out in sera of (1) 47 injured participants (median age, 28 years) collected at baseline (median, 24 days after injury), 3 months, and 2 years, along with the Knee injury and Osteoarthritis Outcome Score, and (2) age- and sex-matched controls. RESULTS Levels of the omega-3 polyunsaturated fatty acids eicosapentaenoic acid (P≤ .0001) and docosahexaenoic acid (P≤ .0001) and the pro-resolving lipid mediators 17- and 14-hydroxydocosahexaenoic acid, and 18-hydroxyeicosapentaenoic acid were all significantly greater at baseline in injured participants compared with the later time points and also higher than in healthy controls (P = .0019 and P≤ .0001, respectively). Levels of pro-inflammatory prostaglandins E2 and D2, leukotriene B4, and thromboxane B2 were significantly lower at baseline compared with the later time points. Higher levels of 8,9-, 11,12-, and 14,15-dihydroxyeicosatrienoic acid (DHET) were cross-sectionally associated with more severe knee pain/symptoms according to the Knee injury and Osteoarthritis Outcome Score at 2 years (P = .0004, R2 = 0.251; P = .0002, R2 = 0.278; and P = .0012, R2 = 0.214, respectively). CONCLUSION The profile of pro-resolving versus pro-inflammatory lipids at baseline suggests an initial activation of pro-resolution pathways, followed by the later activation of pro-inflammatory pathways. CLINICAL RELEVANCE In this largely surgically managed cohort, the association of soluble epoxide hydrolase metabolites, the DHETs, with more severe knee symptoms at 2 years provides a rationale for further investigation into the role of this pathway in persisting knee symptoms in this population, including potential therapeutic strategies.
Collapse
Affiliation(s)
- James Turnbull
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, UK
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Rakesh R. Jha
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - David A. Barrett
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Ana M. Valdes
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
- Injury, Recovery, and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jennifer Alderson
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Williams
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Fortius Clinic, London, UK
| | - Tonia L. Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Fiona E. Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Victoria Chapman
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
- School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
13
|
Giordano R, Ghafouri B, Arendt-Nielsen L, Petersen KKS. Inflammatory biomarkers in patients with painful knee osteoarthritis: exploring the potential link to chronic postoperative pain after total knee arthroplasty-a secondary analysis. Pain 2024; 165:337-346. [PMID: 37703399 DOI: 10.1097/j.pain.0000000000003042] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/19/2023] [Indexed: 09/15/2023]
Abstract
ABSTRACT Total knee arthroplasty (TKA) is the end-stage treatment of knee osteoarthritis (OA), and approximately 20% of patients experience chronic postoperative pain. Studies indicate that inflammatory biomarkers might be associated with pain in OA and potentially linked to the development of chronic postoperative pain after TKA. This study aimed to (1) evaluate preoperative serum levels of inflammatory biomarkers in patients with OA and healthy control subjects, (2) investigate preoperative differences of inflammatory biomarker profiles in subgroups of patients, and (3) compare subgroups of patients with and without postoperative pain 12 months after surgery. Serum samples from patients with OA scheduled for TKA (n = 127) and healthy participants (n = 39) were analyzed. Patients completed the Knee-injury-and-Osteoarthritis-Outcome-Score (KOOS) questionnaire and rated their clinical pain intensity using a visual analog scale (VAS) before and 12 months after TKA. Hierarchical cluster analysis and Orthogonal Partial Least Squares Discriminant Analysis were used to compare groups (patients vs control subjects) and to identify subgroups of patients in relation to postoperative outcomes. Difference in preoperative and postoperative VAS and KOOS scores were compared across subgroups. Twelve inflammatory markers were differentially expressed in patients when compared with control subjects. Cluster analysis identified 2 subgroups of patients with 23 proteins being significantly different ( P < 0.01). The 12-months postoperative VAS and KOOS scores were significantly different between subgroups of patients ( P < 0.05). This study identified differences in specific inflammatory biomarker profiles when comparing patients with OA and control subjects. Cluster analysis identified 2 subgroups of patients with OA, with one subgroup demonstrating comparatively worse 12-month postoperative pain intensity and function scores.
Collapse
Affiliation(s)
- Rocco Giordano
- Center for Neuroplasticity and Pain (CNAP), SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Gistrup, Denmark
- Department of Oral and Maxillofacial Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Bijar Ghafouri
- Pain and Rehabilitation Centre, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Lars Arendt-Nielsen
- Center for Neuroplasticity and Pain (CNAP), SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Gistrup, Denmark
- Center for Mathematical Modeling of Knee Osteoarthritis (MathKOA), Department of Material and Production, Faculty of Engineering and Science, Aalborg University, Aalborg, Denmark
- Department of Gastroenterology & Hepatology, Mech-Sense, Aalborg University Hospital, Aalborg, Denmark
- Steno Diabetes Center North Denmark, Clinical Institute, Aalborg University Hospital, Aalborg, Denmark
| | - Kristian Kjær-Staal Petersen
- Center for Neuroplasticity and Pain (CNAP), SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Gistrup, Denmark
- Center for Mathematical Modeling of Knee Osteoarthritis (MathKOA), Department of Material and Production, Faculty of Engineering and Science, Aalborg University, Aalborg, Denmark
| |
Collapse
|
14
|
Du X, Liu Z, Tao X, Mei Y, Zhou D, Cheng K, Gao S, Shi H, Song C, Zhang X. Research Progress on the Pathogenesis of Knee Osteoarthritis. Orthop Surg 2023; 15:2213-2224. [PMID: 37435789 PMCID: PMC10475681 DOI: 10.1111/os.13809] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 07/13/2023] Open
Abstract
Knee osteoarthritis (KOA) is a chronic joint bone disease characterized by inflammatory destruction and hyperplasia of bone. Its main clinical symptoms are joint mobility difficulties and pain, severe cases can lead to limb paralysis, which poses major pressure to the quality of life and mental health of patients, but also brings serious economic burden to society. The occurrence and development of KOA is influenced by many factors, including systemic factors and local factors. The joint biomechanical changes caused by aging, trauma and obesity, abnormal bone metabolism caused by metabolic syndrome, the effects of cytokines and related enzymes, genetic and biochemical abnormalities caused by plasma adiponectin, etc. all directly or indirectly lead to the occurrence of KOA. However, there is little literature that systematically and comprehensively integrates macro- and microscopic KOA pathogenesis. Therefore, it is necessary to comprehensively and systematically summarize the pathogenesis of KOA in order to provide a better theoretical basis for clinical treatment.
Collapse
Affiliation(s)
- Xin Du
- Center for Phenomics of Traditional Chinese MedicineThe Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Zi‐yu Liu
- Center for Phenomics of Traditional Chinese MedicineThe Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Xing‐xing Tao
- Center for Phenomics of Traditional Chinese MedicineThe Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Yong‐liang Mei
- Department of Orthopaedics and Traumatology (Trauma and Bone‐setting)The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Da‐qian Zhou
- Department of Orthopaedics and Traumatology (Trauma and Bone‐setting)The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Kang Cheng
- Department of Orthopaedics and Traumatology (Trauma and Bone‐setting)The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Si‐long Gao
- Department of Orthopaedics and Traumatology (Trauma and Bone‐setting)The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Hou‐yin Shi
- Medical DepartmentThe Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Chao Song
- Department of Orthopaedics and Traumatology (Trauma and Bone‐setting)The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Xiao‐min Zhang
- Department of Orthopaedics and Traumatology (Trauma and Bone‐setting)The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
15
|
Tasci O, Dogan K. Evaluation of tumour necrosis factor alpha-stimulated gene-6 and fibroblast growth factor-2 levels in patients diagnosed with multi-system inflammatory syndrome in children. Cardiol Young 2023; 33:1086-1091. [PMID: 36918343 DOI: 10.1017/s1047951123000355] [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] [Indexed: 03/16/2023]
Abstract
Investigations are still ongoing about the pathophysiology of multi-system inflammatory syndrome in children, which can progress with serious morbidity and mortality after COVID-19 infection. In this study, we aimed to investigate whether fibroblast growth factor-2 and tumour necrosis factor alpha-stimulated gene-6 levels play a role in the diagnosis of the disease and on cardiac involvement. Twenty-three patients (11 girls, 12 boys) and 26 healthy controls (10 girls, 16 boys) were included in the study. The mean age of the patient and control group was 8.45 ± 2.43 and 10.73 ± 4.27 years, respectively. There was no difference between the fibroblast growth factor-2 and tumour necrosis factor alpha-stimulated gene-6 levels of the patient and control groups. When the patients with myocardial involvement in the patient group were compared with the patients without myocardial involvement in terms of fibroblast growth factor-2 and tumour necrosis factor alpha-stimulated gene-6 levels, no difference was found between these groups. The correlation of fibroblast growth factor-2 and tumour necrosis factor alpha-stimulated gene-6 levels with other laboratory parameters was investigated in the patient group. Fibroblast growth factor-2 was moderately inversely correlated with white blood cell count (r = -0.541, p = 0.008), absolute neutrophil count (r = -0.502, p = 0.015) and C-reactive protein (r = -0.528, p = 0.010). Fibroblast growth factor-2 was strongly inversely correlated with erythrocyte sedimentation rate (r = -0.694, p =<0.001). Our data show that fibroblast growth factor-2 and tumour necrosis factor alpha stimulated gene-6 do not provide sufficient information about diagnosis and cardiac involvement in multi-system inflammatory syndrome in children.
Collapse
Affiliation(s)
- Onur Tasci
- Sivas Numune Hospital, Department of Pediatric Cardiology, Sivas, Turkey
| | - Kubra Dogan
- Sivas Numune Hospital, Department of Biochemistry, Sivas, Turkey
| |
Collapse
|
16
|
Nieboer MF, Reijman M, Wesdorp MA, Bastiaansen-Jenniskens YM, Meuffels DE. Improved Understanding of the Inflammatory Response in Synovial Fluid and Serum after Traumatic Knee Injury, Excluding Fractures of the Knee: A Systematic Review. Cartilage 2023; 14:198-209. [PMID: 36661182 PMCID: PMC10416200 DOI: 10.1177/19476035221141417] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Traumatic knee injury results in a 4- to 10-fold increased risk of post-traumatic osteoarthritis (PTOA). Currently, there are no successful interventions for preventing PTOA after knee injury. The aim of this study is to identify inflammatory proteins that are increased in serum and synovial fluid after acute knee injury, excluding intra-articular fractures. METHODS A literature search was done according to the PRISMA guidelines. Articles reporting about inflammatory proteins after knee injury, except fractures, up to December 8, 2021 were collected. Inclusion criteria were as follows: patients younger than 45 years, no radiographic signs of knee osteoarthritis at baseline, and inflammatory protein measurement within 1 year after trauma. Risk of bias was assessed of the included studies. The level of evidence was determined by the Strength of Recommendation Taxonomy. RESULTS Ten studies were included. All included studies used a healthy control group or the contralateral knee as healthy control. Strong evidence for interleukin 6 (IL-6) and limited evidence for CCL4 show elevated concentrations of these proteins in synovial fluid (SF) after acute knee injury; no upregulation in SF for IL-2, IL-10, CCL3, CCL5, CCL11, granulocyte colony-stimulating factor (G-CSF), and granulocyte-macrophage colony-stimulating factor (GM-CSF) was found. Limited evidence was found for no difference in serum concentration of IL-1β, IL-6, IL-10, CCL2, and tumor necrosis factor alpha (TNF-α) after knee injury. CONCLUSION Interleukin 6 and CCL4 are elevated in SF after acute knee injury. Included studies failed to demonstrate increased concentration of inflammatory proteins in SF samples taken 6 weeks after trauma. Future research should focus on SF inflammatory protein measurements taken less than 6 weeks after injury.
Collapse
Affiliation(s)
- Michael F. Nieboer
- Department of Orthopaedics and Sports Medicine, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Max Reijman
- Department of Orthopaedics and Sports Medicine, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marinus A. Wesdorp
- Department of Orthopaedics and Sports Medicine, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Duncan E. Meuffels
- Department of Orthopaedics and Sports Medicine, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
17
|
Rai MF, Cai L, Zhang Q, Townsend RR, Brophy RH. Synovial Fluid Proteomics From Serial Aspirations of ACL-Injured Knees Identifies Candidate Biomarkers. Am J Sports Med 2023:3635465231169526. [PMID: 37191559 DOI: 10.1177/03635465231169526] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
BACKGROUND Anterior cruciate ligament (ACL) tears often result in knee effusion and an increased risk for developing knee osteoarthritis (OA) in the long run. The molecular profile of these effusions could be informative regarding initial steps in the development of posttraumatic OA after an ACL tear. HYPOTHESIS The proteomics of knee synovial fluid changes over time after ACL injury. STUDY DESIGN Descriptive laboratory study. METHODS Synovial fluid was collected from patients with an acute traumatic ACL tear presenting to the office for evaluation (18.31 ± 19.07 days from injury) (aspiration 1) and again at the time of surgery (35.41 ± 58.15 days after aspiration 1 (aspiration 2). High-resolution liquid chromatography mass spectrometry was used to assess the quantitative protein profile of synovial fluid, and differences in protein profile between the 2 aspirations were determined computationally. RESULTS A total of 58 synovial fluid samples collected from 29 patients (12 male, 17 female; 12 isolated ACL tear, 17 combined ACL and meniscal tear) with a mean age and body mass index of 27.01 ± 12.78 years and 26.30 ± 4.93, respectively, underwent unbiased proteomics analysis. The levels of 130 proteins in the synovial fluid changed over time (87 high, 43 low). Proteins of interest that were significantly higher in aspiration 2 included CRIP1, S100A11, PLS3, POSTN, and VIM, which represent catabolic/inflammatory activities in the joint. Proteins with a known role in chondroprotection and joint homeostasis such as CHI3L2 (YKL-39), TNFAIP6/TSG6, DEFA1, SPP1, and CILP were lower in aspiration 2. CONCLUSION Synovial fluid from knees with ACL tears exhibits an increased burden of inflammatory (catabolic) proteins relevant to OA with reduced levels of chondroprotective (anabolic) proteins. CLINICAL RELEVANCE This study identified a set of novel proteins that provide new biological insights into the aftermath of ACL tears. Elevated inflammation and decreased chondroprotection could represent initial disruption of homeostasis, potentially initiating the development of OA. Longitudinal follow-up and mechanistic studies are necessary to assess the functional role of these proteins in the joint. Ultimately, these investigations could lead to better approaches to predict and possibly improve patient outcomes.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lei Cai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qiang Zhang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - R Reid Townsend
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert H Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
18
|
Damyanovich AZ, Avery L, Staples JR, Marshall KW. 1H NMR Metabolic Profiling of Synovial Fluid from Patients with Anterior Cruciate Ligament Tears and Hemarthrosis. Osteoarthritis Cartilage 2023:S1063-4584(23)00757-4. [PMID: 37146959 DOI: 10.1016/j.joca.2023.03.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/22/2023] [Accepted: 03/29/2023] [Indexed: 05/07/2023]
Abstract
OBJECTIVE To compare the metabolic profiles of synovial fluid (SF) from patients with anterior cruciate ligament tears and hemarthrosis (HA) with that of normal controls, using 1H NMR spectroscopy (NMRS). METHODS Synovial fluid was collected from eleven patients undergoing arthroscopic debridement within fourteen days following an anterior cruciate ligament (ACL) tear and hemarthrosis. Ten additional SF samples were obtained from the knees of osteoarthritis-free volunteers to serve as normal controls. The relative concentrations of twenty-eight endogenous SF metabolites (hydroxybutyrate, acetate, acetoacetate, acetone, alanine, arginine, choline, citrate, creatine, creatinine, formate, glucose, glutamate, glutamine, glycerol, glycine, histidine, isoleucine, lactate, leucine, lysine, phenylalanine, proline, pyruvate, threonine, tyrosine, valine, and the mobile components of glycoproteins and lipids) were evaluated using NMRS and quantified using CHENOMX metabolomics analysis software. Mean differences between groups were evaluated with t-tests controlling for multiple comparisons at an overall error rate of 0.10. RESULTS Statistically significant increases in the levels of glucose, choline, the branched-chain amino acids leucine, isoleucine, and valine, and the mobile components of N-acetyl glycoproteins and lipids were observed in ACL/HA SF as compared with normal controls; lactate levels were reduced. CONCLUSIONS Marked changes occur in the metabolic profiles of human knee fluid following ACL injury and hemarthrosis, suggestive of increased demand and accompanying inflammatory response; potentially increased lipid and glucose metabolism; and possible hyaluronan degradation within the joint following trauma.
Collapse
Affiliation(s)
- Andrei Z Damyanovich
- Department of Medical Physics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Techna Institute, Toronto, Ontario, Canada.
| | - Lisa Avery
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - James R Staples
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - K Wayne Marshall
- Schroeder Arthritis Institute, Division of Orthopaedic Surgery, University Health Network, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Karateev AE, Chernikova AA, Makarov MA. Post-traumatic osteoarthritis: epidemiology, pathogenesis, clinical picture, approaches to pharmacotherapy. MODERN RHEUMATOLOGY JOURNAL 2023. [DOI: 10.14412/1996-7012-2023-1-108-116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Post-traumatic osteoarthritis (PTOA) is an inflammatory and degenerative disease that occurs as a result of the joint structures injury. It is a common pathology, accounting for approximately 12% of all cases of osteoarthritis (OA). PTOA often occurs in people of young productive age, progresses rapidly, causing chronic pain and increasing dysfunction. Individuals undergoing joint replacement for PTOA are, on average, 10 years younger than those with primary OA. The time interval from the moment of injury to the onset of typical PTOA radiological signs varies widely – from 1 year to 15–20 years.The main injuries that cause PTOA are intra-articular fractures, anterior cruciate ligament injuries, meniscus rupture and dislocation of the patella of the knee joint, joint dislocations with damage to the ligamentous apparatus of the ankle and shoulder joints.The pathogenesis of PTOA is determined by chronic inflammation accompanied by macrophage activation, hyperproduction of cytokines, primarily interleukin (IL) 1â, chemokines and growth factors, progressive destruction of joint tissue and degenerative changes (fibrosis, neoangiogenesis, osteophytosis).Pathogenetic treatment of PTOA, which would stop the progression of the disease, has not been developed. The possibility of using inhibitors of IL1â, IL6, inhibitors of tumor necrosis factor á, glucocorticoids, hyaluronic acid, autologous cell based therapy is under study. The control of pain and inflammation in PTOA requires the prescription of traditional drugs that are widely used in the practice of managing patients with primary OA. In particular, the use of symptomatic delayed-acting agents, such as the injectable form of chondroitin sulfate, seems to be appropriate.
Collapse
|
20
|
Sullivan B, Stone AV, Conley CEW, Hunt ER, Lattermann C, Jacobs CA. Human synovial fluid interleukin-6, but not type II collagen breakdown, positively correlated with pain after anterior cruciate ligament injury and reconstruction. J Orthop Res 2023; 41:300-306. [PMID: 35488724 PMCID: PMC9617804 DOI: 10.1002/jor.25355] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023]
Abstract
Anterior cruciate ligament (ACL) injury initiates a biochemical cascade thought to contribute to the onset and progression of posttraumatic osteoarthritis (PTOA). Interleukin-1ß (IL-1ß), IL-6, and C-telopeptide fragments of type II collagen (CTX-II) are implicated in joint inflammation and cartilage degradation following ACL injury; however, their association with pain is still being explored. The purpose of this study was to evaluate the associations between synovial fluid concentrations of IL-1ß, IL-6, and CTX-II with pain following ACL injury and reconstruction. We hypothesized that greater IL-1ß, IL-6, and CTX-II would correlate with greater Pain Visual Analogue Scale (VAS) scores. This was a secondary analysis of 23 patients (mean age = 18.4 years, BMI = 27.4, 13 females/10 males) with acute ACL tears who participated in a pilot randomized trial. Synovial fluid and VAS scores were collected on the day of initial presentation, at ACL reconstruction, and 1 and 4 weeks after surgery. Synovial fluid concentrations of IL-1ß, IL-6, and CTX-II were assessed using enzyme-linked immunoabsorbent assays, and repeated measures correlations were used to assess the relationships between pain and synovial IL-1ß, IL-6, or CTX-II after ACL injury and reconstruction. Pain was positively correlated with synovial fluid IL-6 concentrations (r = 0.52, p < 0.001); however, pain was inversely correlated with CTX-II (r = -0.39, p = 0.002). IL-1ß had no significant correlation with pain. Statement of clinical relevance: PTOA has been described as a "silent killer" and these results suggest that early PTOA may have pro-inflammatory pathways that are not primarily associated with pain but still lead to progressive cartilage loss.
Collapse
Affiliation(s)
- Breanna Sullivan
- Department of Orthopaedic Surgery and Sports Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Austin V Stone
- Department of Orthopaedic Surgery and Sports Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Caitlin E W Conley
- Department of Orthopaedic Surgery and Sports Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Emily R Hunt
- Department of Orthopaedic Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Christian Lattermann
- Department of Orthopaedic Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Cale A Jacobs
- Department of Orthopaedic Surgery and Sports Medicine, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
21
|
Zhu L, Kamalathevan P, Koneva LA, Zarebska JM, Chanalaris A, Ismail H, Wiberg A, Ng M, Muhammad H, Walsby-Tickle J, McCullagh JSO, Watt FE, Sansom SN, Furniss D, Gardiner MD, Vincent TL, Riley N, Spiteri M, McNab I, Little C, Cogswell L, Critchley P, Giele H, Shirley R. Variants in ALDH1A2 reveal an anti-inflammatory role for retinoic acid and a new class of disease-modifying drugs in osteoarthritis. Sci Transl Med 2022; 14:eabm4054. [PMID: 36542696 DOI: 10.1126/scitranslmed.abm4054] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
More than 40% of individuals will develop osteoarthritis (OA) during their lifetime, yet there are currently no licensed disease-modifying treatments for this disabling condition. Common polymorphic variants in ALDH1A2, which encodes the key enzyme for synthesis of all-trans retinoic acid (atRA), are associated with severe hand OA. Here, we sought to elucidate the biological significance of this association. We first confirmed that ALDH1A2 risk variants were associated with hand OA in the U.K. Biobank. Articular cartilage was acquired from 33 individuals with hand OA at the time of routine hand OA surgery. After stratification by genotype, RNA sequencing was performed. A reciprocal relationship between ALDH1A2 mRNA and inflammatory genes was observed. Articular cartilage injury up-regulated similar inflammatory genes by a process that we have previously termed mechanoflammation, which we believe is a primary driver of OA. Cartilage injury was also associated with a concomitant drop in atRA-inducible genes, which were used as a surrogate measure of cellular atRA concentration. Both responses to injury were reversed using talarozole, a retinoic acid metabolism blocking agent (RAMBA). Suppression of mechanoflammation by talarozole was mediated by a peroxisome proliferator-activated receptor gamma (PPARγ)-dependent mechanism. Talarozole was able to suppress mechano-inflammatory genes in articular cartilage in vivo 6 hours after mouse knee joint destabilization and reduced cartilage degradation and osteophyte formation after 26 days. These data show that boosting atRA suppresses mechanoflammation in the articular cartilage in vitro and in vivo and identifies RAMBAs as potential disease-modifying drugs for OA.
Collapse
Affiliation(s)
- Linyi Zhu
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Pragash Kamalathevan
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Lada A Koneva
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Jadwiga Miotla Zarebska
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Anastasios Chanalaris
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Heba Ismail
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
- Healthy Lifespan Institute (HELSI) and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Akira Wiberg
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Michael Ng
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Hayat Muhammad
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - John Walsby-Tickle
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - James S O McCullagh
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Fiona E Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| | - Stephen N Sansom
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Dominic Furniss
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Matthew D Gardiner
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Willcockson H, Ozkan H, Arbeeva L, Mucahit E, Musawwir L, Longobardi L. Early ablation of Ccr2 in aggrecan-expressing cells following knee injury ameliorates joint damage and pain during post-traumatic osteoarthritis. Osteoarthritis Cartilage 2022; 30:1616-1630. [PMID: 36075514 PMCID: PMC9671864 DOI: 10.1016/j.joca.2022.08.015] [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: 11/12/2021] [Revised: 08/10/2022] [Accepted: 08/26/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate whether Ccr2 inactivation in aggrecan-expressing cells induced before post-traumatic OA (PTOA) onset or during progression, improves joint structures, synovial thickness and pain. DESIGN We induced a Ccr2 deletion in aggrecan-expressing cells (CCR2-AggKO) in skeletally mature mice using a tamoxifen-inducible Ccr2 inactivation. We stimulated PTOA changes (destabilization of medial meniscus, DMM) in CCR2-AggKO and CCR2+/+ mice, inducing recombination before DMM or 4 wks after DMM (early-vs late-inactivation). Joint damage was evaluated 2, 4, 8, 12 wks post-DMM using multiple scores: articular-cartilage structure (ACS), Safranin-O, histomorphometry, osteophyte size/maturity, subchondral bone thickness and synovial hyperplasia. Spontaneous (incapacitance meter) and evoked pain (von-Frey filaments) were assessed up to 20 wks. RESULTS Early aggrecan-Ccr2 inactivation in CCR2-AggKO mice (N=8) resulted in improved ACS score (8-12wk, P=0.002), AC area (4-12wk, P<0.05) and Saf-O score (2wks P=0.004, 4wks P=0.02, 8-12wks P=0.002) compared to CCR2+/+. Increased subchondral bone thickness was delayed only at 2 wks and exclusively following early recombination. Osteophyte size was not affected, but osteophyte maturation (cartilage-to-bone) was delayed (4wks P=0.04; 8 wks P=0.03). Although late aggrecan-Ccr2 deletion led to some cartilage improvement, most data did not reach statistical significance; osteophyte maturity was delayed at 12wks. Early aggrecan-Ccr2 deletion led to improved pain measures of weight bearing compared to CCR2+/+ mice (N = 9, 12wks diff 0.13 [0.01, 0.26], 16wks diff 0.15 [0.05, 0.26], 20wks diff 0.23 [0.14, 0.31]). Improved mechanosensitivity in evoked pain, although less noticeable, was detected. CONCLUSIONS We demonstrated that deletion of Ccr2 in aggrecan expressing cells reduces the initiation but not progression of OA.
Collapse
Affiliation(s)
- H Willcockson
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - H Ozkan
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - L Arbeeva
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - E Mucahit
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - L Musawwir
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - L Longobardi
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| |
Collapse
|
23
|
Aman ZS, DePhillipo NN, Familiari F, Dickens JF, LaPrade RF, Dekker TJ. Acute Intervention With Selective Interleukin-1 Inhibitor Therapy May Reduce the Progression of Posttraumatic Osteoarthritis of the Knee: A Systematic Review of Current Evidence. Arthroscopy 2022; 38:2543-2556. [PMID: 35189307 DOI: 10.1016/j.arthro.2022.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE To evaluate the efficacy of selective interleukin (IL)-1 inhibitor therapy in the reduction of posttraumatic osteoarthritis (PTOA) progression following knee ligament or meniscal injury. METHODS A systematic review was conducted evaluating the disease-modifying efficacy of selective IL-1 inhibition in the setting of knee PTOA. RESULTS The literature search identified 364 articles and 11 studies were included (n = 10 preclinical, n = 1 clinical). Drug delivery in preclinical studies was administered using IL-1Ra-encoded helper-dependent adenovirus particles (n = 3), synovial cells transfected with an IL-1Ra-encoded retroviral vector (n = 3), or varying chemical compositions of nonviral microcapsule gene carriers (n = 4). Intervention with selective IL-1 inhibitor therapy within 2 weeks of injury provided the greatest protective benefits in reducing the progression of PTOA regardless of drug delivery methodology in preclinical models. The majority of studies reported significantly better cartilage integrity and reduction in lesion size in animals treated with gene therapy with the greatest effects seen in those treated within 5 to 7 days of injury. CONCLUSIONS Early intervention with selective IL-1 inhibitor therapy were effective in reducing proinflammatory IL-1β levels in the acute and subacute phases following traumatic knee injury in preclinical animal model studies, while significantly reducing cartilage damage, lesion size, and PTOA progression at short-term follow-up. However, it was found that the effect of these therapies diminished over time. CLINICAL RELEVANCE Acute, intra-articular injection of selective IL-1 inhibitors may reduce PTOA progression, supporting the need for additional basic and clinical investigation.
Collapse
Affiliation(s)
- Zachary S Aman
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, U.S.A
| | | | - Filippo Familiari
- Department of Orthopaedics and Trauma Surgery, Magna Graecia University, Catanzaro, Italy
| | | | | | | |
Collapse
|
24
|
Keil LG, Onuscheck DS, Pratson LF, Kamath GV, Creighton RA, Nissman DB, Pietrosimone BG, Spang JT. Bone bruising severity after anterior cruciate ligament rupture predicts elevation of chemokine MCP-1 associated with osteoarthritis. J Exp Orthop 2022; 9:37. [PMID: 35476154 PMCID: PMC9046516 DOI: 10.1186/s40634-022-00478-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/16/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose Anterior cruciate ligament rupture is associated with characteristic bone contusions in approximately 80% of patients, and these have been correlated with higher pain scores. Bone bruising may indicate joint damage that increases inflammation and the likelihood of posttraumatic osteoarthritis. We sought to characterize the severity of bone bruising following acute anterior cruciate ligament injury and determine if it correlates with synovial fluid and serum levels of the proinflammatory chemokine monocyte chemoattractant protein-1 associated with posttraumatic osteoarthritis. Methods This was a retrospective analysis of data collected prospectively from January 2014 through December 2016. All patients who sustained an acute ligament rupture were evaluated within 15 days of injury, obtained a magnetic resonance imaging study, and underwent bone-patellar-tendon-bone autograft reconstruction were offered enrollment. The overall severity of bone bruising on magnetic resonance imaging was graded (sum of 0–3 grades in 13 sectors of the articular surfaces). Serum and synovial fluid levels of monocyte chemoattractant protein-1 were measured within 14 days of injury, and serum levels were again measured 6 and 12 months following surgery. Separate univariate linear regression models were constructed to determine the association between monocyte chemoattractant protein-1 and bone bruising severity at each time point. Results Forty-eight subjects were included in this study. They had a mean age of 21.4 years and were 48% female. Median overall bone bruising severity was 5 (range 0–14). Severity of bone bruising correlated with higher synovial fluid concentrations of monocyte chemoattractant protein-1 preoperatively (R2 = 0.18, p = 0.009) and with serum concentrations at 12 months post-reconstruction (R2 = 0.12, p = 0.04). Conclusions The severity of bone bruising following anterior cruciate ligament rupture is associated with higher levels of the proinflammatory cytokine monocyte chemoattractant protein-1 in synovial fluid acutely post-injury and in serum 12-months following anterior cruciate ligament reconstruction. This suggests that severe bone bruising on magnetic resonance imaging after ligament rupture may indicate increased risk for persistent joint inflammation and posttraumatic osteoarthritis. Level of evidence III ― retrospective cohort study.
Collapse
Affiliation(s)
- Lukas G Keil
- Department of Orthopaedic Surgery, School of Medicine, University of North Carolina, 130 Mason Farm Road, CB# 7055, Chapel Hill, NC, 27599-7055, USA.
| | | | - Lincoln F Pratson
- Department of Orthopaedic Surgery, School of Medicine, University of North Carolina, 130 Mason Farm Road, CB# 7055, Chapel Hill, NC, 27599-7055, USA
| | - Ganesh V Kamath
- Department of Orthopaedic Surgery, School of Medicine, University of North Carolina, 130 Mason Farm Road, CB# 7055, Chapel Hill, NC, 27599-7055, USA
| | - Robert A Creighton
- Department of Orthopaedic Surgery, School of Medicine, University of North Carolina, 130 Mason Farm Road, CB# 7055, Chapel Hill, NC, 27599-7055, USA
| | - Daniel B Nissman
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Brian G Pietrosimone
- Department of Exercise and Sport Science, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffrey T Spang
- Department of Orthopaedic Surgery, School of Medicine, University of North Carolina, 130 Mason Farm Road, CB# 7055, Chapel Hill, NC, 27599-7055, USA
| |
Collapse
|
25
|
Chaudhry N, Muhammad H, Seidl C, Downes D, Young DA, Hao Y, Zhu L, Vincent TL. Highly efficient CRISPR-Cas9-mediated editing identifies novel mechanosensitive microRNA-140 targets in primary human articular chondrocytes. Osteoarthritis Cartilage 2022; 30:596-604. [PMID: 35074547 PMCID: PMC8987936 DOI: 10.1016/j.joca.2022.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE MicroRNA 140 (miR-140) is a chondrocyte-specific endogenous gene regulator implicated in osteoarthritis (OA). As mechanical injury is a primary aetiological factor in OA, we investigated miR-140-dependent mechanosensitive gene regulation using a novel CRISPR-Cas9 methodology in primary human chondrocytes. METHOD Primary (passage 1/2) human OA chondrocytes were isolated from arthroplasty samples (six donors) and transfected with ribonuclear protein complexes or plasmids using single guide RNAs (sgRNAs) targeting miR-140, in combination with Cas9 endonuclease. Combinations of sgRNAs and single/double transfections were tested. Gene editing was measured by T7 endonuclease 1 (T7E1) assay. miRNA levels were confirmed by qPCR in chondrocytes and in wild type murine femoral head cartilage after acute injury. Predicted close match off-targets were examined. Mechanosensitive miR-140 target validation was assessed in 42 injury-associated genes using TaqMan Microfluidic cards in targeted and donor-matched control chondrocytes. Identified targets were examined in RNAseq data from costal chondrocytes from miR-140-/- mice. RESULTS High efficiency gene editing of miR-140 (90-98%) was obtained when two sgRNAs were combined with double RNP-mediated CRISPR-Cas9 transfection. miR-140 levels fell rapidly after femoral cartilage injury. Of the top eight miR-140 gene targets identified (P < 0.01), we validated three previously identified ones (septin 2, bone morphogenetic protein 2 and fibroblast growth factor 2). Novel targets included Agrin, a newly recognised pro-regenerative cartilage agent, and proteins associated with retinoic acid signalling and the primary cilium. CONCLUSION We describe a highly efficient CRISPR-Cas9-mediated strategy for gene editing in primary human chondrocytes and identify several novel mechanosensitive miR-140 targets of disease relevance.
Collapse
Affiliation(s)
- N Chaudhry
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY, United Kingdom
| | - H Muhammad
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY, United Kingdom
| | - C Seidl
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY, United Kingdom
| | - D Downes
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS, United Kingdom
| | - D A Young
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ, United Kingdom
| | - Y Hao
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ, United Kingdom
| | - L Zhu
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY, United Kingdom
| | - T L Vincent
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY, United Kingdom.
| |
Collapse
|
26
|
Vincent TL. Post-traumatic OA - are we any closer to prevention? Osteoarthritis Cartilage 2021; 29:1630-1631. [PMID: 34903334 DOI: 10.1016/j.joca.2021.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 02/02/2023]
Affiliation(s)
- T L Vincent
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, London OX37FY, United Kingdom.
| |
Collapse
|
27
|
Haubruck P, Pinto MM, Moradi B, Little CB, Gentek R. Monocytes, Macrophages, and Their Potential Niches in Synovial Joints - Therapeutic Targets in Post-Traumatic Osteoarthritis? Front Immunol 2021; 12:763702. [PMID: 34804052 PMCID: PMC8600114 DOI: 10.3389/fimmu.2021.763702] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Synovial joints are complex structures that enable normal locomotion. Following injury, they undergo a series of changes, including a prevalent inflammatory response. This increases the risk for development of osteoarthritis (OA), the most common joint disorder. In healthy joints, macrophages are the predominant immune cells. They regulate bone turnover, constantly scavenge debris from the joint cavity and, together with synovial fibroblasts, form a protective barrier. Macrophages thus work in concert with the non-hematopoietic stroma. In turn, the stroma provides a scaffold as well as molecular signals for macrophage survival and functional imprinting: “a macrophage niche”. These intricate cellular interactions are susceptible to perturbations like those induced by joint injury. With this review, we explore how the concepts of local tissue niches apply to synovial joints. We introduce the joint micro-anatomy and cellular players, and discuss their potential interactions in healthy joints, with an emphasis on molecular cues underlying their crosstalk and relevance to joint functionality. We then consider how these interactions are perturbed by joint injury and how they may contribute to OA pathogenesis. We conclude by discussing how understanding these changes might help identify novel therapeutic avenues with the potential of restoring joint function and reducing post-traumatic OA risk.
Collapse
Affiliation(s)
- Patrick Haubruck
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Marlene Magalhaes Pinto
- Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Babak Moradi
- Clinic of Orthopaedics and Trauma Surgery, University Clinic of Schleswig-Holstein, Kiel, Germany
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Rebecca Gentek
- Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
28
|
Meehan RT, Regan EA, Hoffman ED, Wolf ML, Gill MT, Crooks JL, Parmar PJ, Scheuring RA, Hill JC, Pacheco KA, Knight V. Synovial Fluid Cytokines, Chemokines and MMP Levels in Osteoarthritis Patients with Knee Pain Display a Profile Similar to Many Rheumatoid Arthritis Patients. J Clin Med 2021; 10:jcm10215027. [PMID: 34768546 PMCID: PMC8584576 DOI: 10.3390/jcm10215027] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 12/20/2022] Open
Abstract
Background: There are currently no effective disease-modifying drugs to prevent cartilage loss in osteoarthritis and synovial fluid is a potentially valuable source of biomarkers to understand the pathogenesis of different types of arthritis and identify drug responsiveness. The aim of this study was to compare the differences between SF cytokines and other proteins in patients with OA (n = 21) to those with RA (n = 27) and normal knees (n = 3). Methods: SF was obtained using ultrasound (US) guidance and an external pneumatic compression device. RA patients were categorized as active (n = 20) or controlled (n = 7) based upon SF white blood cell counts (> or <300 cells/mm3). Samples were cryopreserved and analyzed by multiplex fluorescent bead assays (Luminex). Between-group differences of 16 separate biomarker proteins were identified using ANOVA on log10-transformed concentrations with p values adjusted for multiple testing. Results: Only six biomarkers were significantly higher in SF from active RA compared to OA—TNF-α, IL-1-β IL-7, MMP-1, MMP-2, and MMP-3. Only MMP-8 levels in RA patients correlated with SF WBC counts (p < 0.0001). Among OA patients, simultaneous SF IL-4, IL-6, IL-8, and IL-15 levels were higher than serum levels, whereas MMP-8, MMP-9, and IL-18 levels were higher in serum (p < 0.05). Conclusion: These results support the growing evidence that OA patients have a pro-inflammatory/catabolic SF environment. SF biomarker analysis using multiplex testing and US guidance may distinguish OA phenotypes and identify treatment options based upon targeted inflammatory pathways similar to patients with RA.
Collapse
Affiliation(s)
- Richard T. Meehan
- Department of Medicines, Immunology Labs and Bioinformatics National Jewish Health, Denver, CO 80206, USA; (E.A.R.); (E.D.H.); (M.L.W.); (M.T.G.); (J.L.C.); (K.A.P.)
- Correspondence:
| | - Elizabeth A. Regan
- Department of Medicines, Immunology Labs and Bioinformatics National Jewish Health, Denver, CO 80206, USA; (E.A.R.); (E.D.H.); (M.L.W.); (M.T.G.); (J.L.C.); (K.A.P.)
| | - Eric D. Hoffman
- Department of Medicines, Immunology Labs and Bioinformatics National Jewish Health, Denver, CO 80206, USA; (E.A.R.); (E.D.H.); (M.L.W.); (M.T.G.); (J.L.C.); (K.A.P.)
| | - Molly L. Wolf
- Department of Medicines, Immunology Labs and Bioinformatics National Jewish Health, Denver, CO 80206, USA; (E.A.R.); (E.D.H.); (M.L.W.); (M.T.G.); (J.L.C.); (K.A.P.)
| | - Mary T. Gill
- Department of Medicines, Immunology Labs and Bioinformatics National Jewish Health, Denver, CO 80206, USA; (E.A.R.); (E.D.H.); (M.L.W.); (M.T.G.); (J.L.C.); (K.A.P.)
| | - James L. Crooks
- Department of Medicines, Immunology Labs and Bioinformatics National Jewish Health, Denver, CO 80206, USA; (E.A.R.); (E.D.H.); (M.L.W.); (M.T.G.); (J.L.C.); (K.A.P.)
- Colorado School of Public Health, CU Anschutz School of Medicine, Aurora, CO 80045, USA
| | - Prashant J. Parmar
- Department of Internal Medicine, National Jewish Health, Saint Joseph Hospital, Denver, CO 80218, USA;
| | | | - John C. Hill
- CU Sports Medicine, Department of Orthopedic Surgery, University of Colorado, Denver, CO 80222, USA;
| | - Karin A. Pacheco
- Department of Medicines, Immunology Labs and Bioinformatics National Jewish Health, Denver, CO 80206, USA; (E.A.R.); (E.D.H.); (M.L.W.); (M.T.G.); (J.L.C.); (K.A.P.)
| | - Vijaya Knight
- Immunology Department, Children’s Hospital, Denver, CO 80045, USA;
| |
Collapse
|
29
|
Garriga C, Goff M, Paterson E, Hrusecka R, Hamid B, Alderson J, Leyland K, Honeyfield L, Greenshields L, Satchithananda K, Lim A, Arden NK, Judge A, Williams A, Vincent TL, Watt FE. Clinical and molecular associations with outcomes at 2 years after acute knee injury: a longitudinal study in the Knee Injury Cohort at the Kennedy (KICK). THE LANCET. RHEUMATOLOGY 2021; 3:e648-e658. [PMID: 34476411 PMCID: PMC8390381 DOI: 10.1016/s2665-9913(21)00116-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Joint injury is a major risk factor for osteoarthritis and provides an opportunity to prospectively examine early processes associated with osteoarthritis. We investigated whether predefined baseline demographic and clinical factors, and protein analytes in knee synovial fluid and in plasma or serum, were associated with clinically relevant outcomes at 2 years after knee injury. METHODS This longitudinal cohort study recruited individuals aged 16-50 years between Nov 1, 2010, and Nov 28, 2014, across six hospitals and clinics in London, UK. Participants were recruited within 8 weeks of having a clinically significant acute knee injury (effusion and structural injury on MRI), which was typically treated surgically. We measured several predefined clinical variables at baseline (eg, time from injury to sampling, extent and type of joint injury, synovial fluid blood staining, presence of effusion, self-reported sex, age, and BMI), and measured 12 synovial fluid and four plasma or serum biomarkers by immunoassay at baseline and 3 months. The primary outcome was Knee Injury and Osteoarthritis Outcome Score (KOOS4) at 2 years, adjusted for baseline score, assessed in all patients. Linear and logistic regression models adjusting for predefined covariates were used to assess associations between baseline variables and 2-year KOOS4. This study is registered with ClinicalTrials.gov, number NCT02667756. FINDINGS We enrolled 150 patients at a median of 17 days (range 1-59, IQR 9-26) after knee injury. 123 (82%) were male, with a median age of 25 years (range 16-50, IQR 21-30). 98 (65%) of 150 participants completed a KOOS4 at 2 (or 3) years after enrolment (50 participants were lost to follow-up and two were withdrawn due to adverse events unrelated to study participation); 77 (51%) participants had all necessary variables available and were included in the core variable adjusted analysis. In the 2-year dataset mean KOOS4 improved from 38 (SD 18) at baseline to 79 (18) at 2 years. Baseline KOOS4, medium-to-large knee effusion, and moderate-to-severe synovial blood staining and their interaction significantly predicted 2-year KOOS4 (n=77; coefficient -20·5, 95% CI -34·8 to -6·18; p=0·0060). The only predefined biomarkers that showed independent associations with 2-year KOOS4 were synovial fluid MCP-1 (n=77; -0·015, 0·027 to -0·004 per change in 1 pg/mL units; p=0·011) and IL-6 (n=77; -0·0005, -0·0009 to -0·0001 per change in 1 pg/mL units; p=0·017). These biomarkers, combined with the interaction of effusion and blood staining, accounted for 39% of outcome variability. Two adverse events occurred that were linked to study participation, both at the time of blood sampling (one presyncopal episode, one tenderness and pain at the site of venepuncture). INTERPRETATION The combination of effusion and haemarthrosis was significantly associated with symptomatic outcomes after acute knee injury. The synovial fluid molecular protein response to acute knee injury (best represented by MCP-1 and IL-6) was independently associated with symptomatic outcomes but not with structural outcomes, with the biomarkers overall playing a minor role relative to clinical predictors. The relationship between symptoms and structure after acute knee injury and their apparent dissociation early in this process need to be better understood to make clinical progress. FUNDING Versus Arthritis, Kennedy Trust for Rheumatology Research, and NIHR Oxford Biomedical Research Centre.
Collapse
Affiliation(s)
- Cesar Garriga
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
- Centre for Statistics in Medicine, NDORMS, University of Oxford, Oxford, UK
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Megan Goff
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Erin Paterson
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Renata Hrusecka
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Benjamin Hamid
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Jennifer Alderson
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Kirsten Leyland
- NIHR Bristol BRC, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Lesley Honeyfield
- Department of Radiology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Liam Greenshields
- Department of Radiology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Keshthra Satchithananda
- Department of Radiology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
- Department of Radiology, King's College Hospital NHS Foundation Trust, London, UK
| | - Adrian Lim
- Department of Radiology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Nigel K Arden
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
- Centre for Sports, Exercise and Osteoarthritis Research Versus Arthritis, NDORMS, University of Oxford, Oxford, UK
| | - Andrew Judge
- Centre for Statistics in Medicine, NDORMS, University of Oxford, Oxford, UK
- NIHR Bristol BRC, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
- Musculoskeletal Research Unit, University of Bristol, Bristol, UK
| | - Andrew Williams
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
- Fortius Clinic, London, UK
| | - Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
- Department of Rheumatology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Fiona E Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
- Centre for Sports, Exercise and Osteoarthritis Research Versus Arthritis, NDORMS, University of Oxford, Oxford, UK
- Department of Rheumatology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
- Department of Immunology and Inflammation, Imperial College London, London, UK
| |
Collapse
|
30
|
Molnar V, Matišić V, Kodvanj I, Bjelica R, Jeleč Ž, Hudetz D, Rod E, Čukelj F, Vrdoljak T, Vidović D, Starešinić M, Sabalić S, Dobričić B, Petrović T, Antičević D, Borić I, Košir R, Zmrzljak UP, Primorac D. Cytokines and Chemokines Involved in Osteoarthritis Pathogenesis. Int J Mol Sci 2021; 22:9208. [PMID: 34502117 PMCID: PMC8431625 DOI: 10.3390/ijms22179208] [Citation(s) in RCA: 283] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/15/2021] [Accepted: 08/24/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis is a common cause of disability worldwide. Although commonly referred to as a disease of the joint cartilage, osteoarthritis affects all joint tissues equally. The pathogenesis of this degenerative process is not completely understood; however, a low-grade inflammation leading to an imbalance between anabolic and katabolic processes is a well-established factor. The complex network of cytokines regulating these processes and cell communication has a central role in the development and progression of osteoarthritis. Concentrations of both proinflammatory and anti-inflammatory cytokines were found to be altered depending on the osteoarthritis stage and activity. In this review, we analyzed individual cytokines involved in the immune processes with an emphasis on their function in osteoarthritis.
Collapse
Affiliation(s)
- Vilim Molnar
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Vid Matišić
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
| | - Ivan Kodvanj
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Roko Bjelica
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
| | - Željko Jeleč
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Nursing, University North, 48000 Varaždin, Croatia
| | - Damir Hudetz
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Orthopaedic Surgery, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Eduard Rod
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
| | - Fabijan Čukelj
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- University Hospital “Sisters of Mercy”, Clinic for Traumatology, Draškovićeva 19, 10000 Zagreb, Croatia;
- Department of Health Studies, University of Split, 21000 Split, Croatia
- Department of Traumatology, Medical University Merkur Hospital, 10000 Zagreb, Croatia
| | - Trpimir Vrdoljak
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Orthopaedic Surgery, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Dinko Vidović
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- University Hospital “Sisters of Mercy”, Clinic for Traumatology, Draškovićeva 19, 10000 Zagreb, Croatia;
| | | | - Srećko Sabalić
- University Hospital “Sisters of Mercy”, Clinic for Traumatology, Draškovićeva 19, 10000 Zagreb, Croatia;
| | - Borut Dobričić
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Orthopaedics and Traumatology, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Tadija Petrović
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Health Studies, University of Split, 21000 Split, Croatia
| | - Darko Antičević
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Igor Borić
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Traumatology, Medical University Merkur Hospital, 10000 Zagreb, Croatia
- Medical School, University of Split, 21000 Split, Croatia;
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
| | - Rok Košir
- Molecular Biology Laboratory, BIA Separations CRO, Labena Ltd., 1000 Ljubljana, Slovenia; (R.K.); (U.P.Z.)
| | - Uršula Prosenc Zmrzljak
- Molecular Biology Laboratory, BIA Separations CRO, Labena Ltd., 1000 Ljubljana, Slovenia; (R.K.); (U.P.Z.)
| | - Dragan Primorac
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Split, 21000 Split, Croatia;
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96450 Coburg, Germany
- Eberly College of Science, State College, The Pennsylvania State University, University Park, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
| |
Collapse
|
31
|
Perry J, Roelofs AJ, Mennan C, McCarthy HS, Richmond A, Clark SM, Riemen AHK, Wright K, De Bari C, Roberts S. Human Mesenchymal Stromal Cells Enhance Cartilage Healing in a Murine Joint Surface Injury Model. Cells 2021; 10:1999. [PMID: 34440768 PMCID: PMC8393840 DOI: 10.3390/cells10081999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 01/15/2023] Open
Abstract
Human umbilical cord (hUC)- or bone marrow (hBM)-derived mesenchymal stromal cells (MSCs) were evaluated as an allogeneic source of cells for cartilage repair. We aimed to determine if they could enhance healing of chondral defects with or without the recruitment of endogenous cells. hMSCs were applied into a focal joint surface injury in knees of adult mice expressing tdTomato fluorescent protein in cells descending from Gdf5-expressing embryonic joint interzone cells. Three experimental groups were used: (i) hUC-MSCs, (ii) hBM-MSCs and (iii) PBS (vehicle) without cells. Cartilage repair was assessed after 8 weeks and tdTomato-expressing cells were detected by immunostaining. Plasma levels of pro-inflammatory mediators and other markers were measured by electrochemiluminescence. Both hUC-MSC (n = 14, p = 0.009) and hBM-MSC (n = 13, p = 0.006) treatment groups had significantly improved cartilage repair compared to controls (n = 18). While hMSCs were not detectable in the repair tissue at 8 weeks post-implantation, increased endogenous Gdf5-lineage cells were detected in repair tissue of hUC-MSC-treated mice. This xenogeneic study indicates that hMSCs enhance intrinsic cartilage repair mechanisms in mice. Hence, hMSCs, particularly the more proliferative hUC-MSCs, could represent an attractive allogeneic cell population for treating patients with chondral defects and perhaps prevent the onset and progression of osteoarthritis.
Collapse
Affiliation(s)
- Jade Perry
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| | - Anke J. Roelofs
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Claire Mennan
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| | - Helen S. McCarthy
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| | - Alison Richmond
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Susan M. Clark
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Anna H. K. Riemen
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Karina Wright
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| | - Cosimo De Bari
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Sally Roberts
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| |
Collapse
|
32
|
Khella CM, Horvath JM, Asgarian R, Rolauffs B, Hart ML. Anti-Inflammatory Therapeutic Approaches to Prevent or Delay Post-Traumatic Osteoarthritis (PTOA) of the Knee Joint with a Focus on Sustained Delivery Approaches. Int J Mol Sci 2021; 22:8005. [PMID: 34360771 PMCID: PMC8347094 DOI: 10.3390/ijms22158005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation plays a central role in the pathogenesis of knee PTOA after knee trauma. While a comprehensive therapy capable of preventing or delaying post-traumatic osteoarthritis (PTOA) progression after knee joint injury does not yet clinically exist, current literature suggests that certain aspects of early post-traumatic pathology of the knee joint may be prevented or delayed by anti-inflammatory therapeutic interventions. We discuss multifaceted therapeutic approaches that may be capable of effectively reducing the continuous cycle of inflammation and concomitant processes that lead to cartilage degradation as well as those that can simultaneously promote intrinsic repair processes. Within this context, we focus on early disease prevention, the optimal timeframe of treatment and possible long-lasting sustained delivery local modes of treatments that could prevent knee joint-associated PTOA symptoms. Specifically, we identify anti-inflammatory candidates that are not only anti-inflammatory but also anti-degenerative, anti-apoptotic and pro-regenerative.
Collapse
Affiliation(s)
| | | | | | | | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs—University of Freiburg, 79085 Freiburg im Breisgau, Germany; (C.M.K.); (J.M.H.); (R.A.); (B.R.)
| |
Collapse
|
33
|
Abstract
OBJECTIVES Osteoarthritis (OA) is known to be a slowly progressive disease that alters all tissue compartments of the joint involved with a characteristic degradation of the cartilage, bone remodeling, and inflammation. One of the prominent symptoms in OA patients is pain, but a few radiologic, inflammatory, or structurally related biomarkers have shown few if any associations with pain. This study aimed to assess serum levels of 92 markers involved in inflammatory pathways in patients with knee osteoarthritis (KOA) and evaluate their possible associations with the clinical pain intensity. MATERIALS AND METHODS Serum samples were collected from 127 KOA patients and 39 healthy participants with no knee pain. Each serum sample was analyzed for 92 inflammatory markers using the Proximity Extension Array (PEA) technology. Clinical pain intensity was assessed using a Visual Analog Scale, and patients completed the Knee Injury and Osteoarthritis Outcome Score (KOOS) questionnaire. RESULTS Fifteen markers were significantly different when comparing KOA patients and healthy participants. Two markers, fibroblast growth factor-21 and Eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), correlated positively with pain intensity (R=0.235, P=0.008; R=0.233, P=0.008). Moreover, a linear regression model showed interleukin-6, macrophage colony-stimulating factor 1, fibroblast growth factor-21, and tumor necrosis factor superfamily member 12 (TWEAK) as significant independent parameters for pain intensity. DISCUSSION The associations between specific cytokines and KOA pain intensities provide new insights into the understanding of the underlying factors driving the pain in OA.
Collapse
|
34
|
Mason D, Englund M, Watt FE. Prevention of posttraumatic osteoarthritis at the time of injury: Where are we now, and where are we going? J Orthop Res 2021; 39:1152-1163. [PMID: 33458863 DOI: 10.1002/jor.24982] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/11/2021] [Indexed: 02/04/2023]
Abstract
This overview of progress made in preventing post-traumatic osteoarthritis (PTOA) was delivered in a workshop at the Orthopaedics Research Society Annual Conference in 2019. As joint trauma is a major risk factor for OA, defining the molecular changes within the joint at the time of injury may enable the targeting of biological processes to prevent later disease. Animal models have been used to test therapeutic targets to prevent PTOA. A review of drug treatments for PTOA in rodents and rabbits between 2016 and 2018 revealed 11 systemic interventions, 5 repeated intra-articular or topical interventions, and 5 short-term intra-articular interventions, which reduced total Osteoarthritis Research Society International scores by 30%-50%, 20%-70%, and 0%-40%, respectively. Standardized study design, reporting of effect size, and quality metrics, alongside a "whole joint" approach to assessing efficacy, would improve the translation of promising new drugs. A roadblock to translating preclinical discoveries has been the lack of guidelines on the design and conduct of human trials to prevent PTOA. An international workshop addressing this in 2016 considered inclusion criteria and study design, and advocated the use of experimental medicine studies to triage candidate treatments and the development of early biological and imaging biomarkers. Human trials for the prevention of PTOA have tested anakinra after anterior cruciate ligament rupture and dexamethasone after radiocarpal injury. PTOA offers a unique opportunity for defining early mechanisms of OA to target therapeutically. Progress in trial design and high-quality preclinical research, and allegiance with patients, regulatory bodies, and the pharmaceutical industry, will advance this field.
Collapse
Affiliation(s)
- Deborah Mason
- Biomechanics and Bioengineeering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | - Martin Englund
- Faculty of Medicine, Department of Clinical Sciences Lund, Orthopedics, Clinical Epidemiology Unit, Lund Unversity, Lund, Sweden
| | - Fiona E Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Hulme CH, Peffers MJ, Harrington GMB, Wilson E, Perry J, Roberts S, Gallacher P, Jermin P, Wright KT. Identification of Candidate Synovial Fluid Biomarkers for the Prediction of Patient Outcome After Microfracture or Osteotomy. Am J Sports Med 2021; 49:1512-1523. [PMID: 33787363 DOI: 10.1177/0363546521995565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Biomarkers are needed to predict clinical outcomes for microfracture and osteotomy surgeries to ensure patients can be better stratified to receive the most appropriate treatment. PURPOSE To identify novel biomarker candidates and to investigate the potential of a panel of protein biomarkers for the prediction of clinical outcome after treatment with microfracture or osteotomy. STUDY DESIGN Descriptive laboratory study. METHODS To identify novel candidate biomarker proteins, we used label-free quantitation after liquid chromatography-tandem mass spectrometry of dynamic range-compressed synovial fluids (SFs) from individuals who responded excellently or poorly (based on change in Lysholm score) to microfracture (n = 6) or osteotomy (n = 7). Biomarkers that were identified in this proteomic analysis or that relate to osteoarthritis (OA) severity or have predictive value in another early OA therapy (autologous cell implantation) were measured in the SF of 19 and 13 patients before microfracture or osteotomy, respectively, using commercial immunoassays, and were normalized to urea. These were aggrecanase-1 (ADAMTS-4), cartilage oligomeric matrix protein (COMP), hyaluronan (HA), lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1), matrix metalloproteinase 1 and 3, soluble CD14, S100 calcium binding protein A13, and 14-3-3 protein theta (YWHAQ). Levels of COMP and HA were also measured in the plasma of these patients. To find predictors of postoperative function, multivariable regression analyses were performed. RESULTS Proteomic analyses highlighted YWHAQ and LYVE-1 as being differentially abundant between the clinical responders/improvers and nonresponders after microfracture. A linear regression model after backward variable selection could relate preoperative concentrations of SF proteins (HA, YWHAQ, LYVE-1), activity of ADAMTS-4, and patient demographic characteristics (smoker status and sex) with Lysholm score 12 months after microfracture. Further, a generalized linear model with elastic net penalization indicated that lower preoperative activity of ADAMTS-4 in SF, being a nonsmoker, and being younger at the time of operation were indicative of a higher postoperative Lysholm score (improved joint function) after osteotomy surgery. CONCLUSION We have identified biomarkers and generated regression models with the potential to predict clinical outcome in patients treated with microfracture or osteotomy of the knee. CLINICAL RELEVANCE Candidate protein biomarkers identified in this study have the potential to help determine which patients will be best suited to treatment with microfracture or osteotomy.
Collapse
Affiliation(s)
- Charlotte H Hulme
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, UK
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Mandy J Peffers
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Gabriel Mateus Bernardo Harrington
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, UK
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Emma Wilson
- Chester Medical School, Chester University, Chester, UK
| | - Jade Perry
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, UK
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Sally Roberts
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, UK
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Pete Gallacher
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Paul Jermin
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Karina T Wright
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, UK
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| |
Collapse
|
36
|
Ishihara S, Obeidat AM, Wokosin DL, Ren D, Miller RJ, Malfait AM, Miller RE. The role of intra-articular neuronal CCR2 receptors in knee joint pain associated with experimental osteoarthritis in mice. Arthritis Res Ther 2021; 23:103. [PMID: 33827672 PMCID: PMC8025346 DOI: 10.1186/s13075-021-02486-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background C–C chemokine receptor 2 (CCR2) signaling plays a key role in pain associated with experimental murine osteoarthritis (OA) after destabilization of the medial meniscus (DMM). Here, we aimed to assess if CCR2 expressed by intra-articular sensory neurons contributes to knee hyperalgesia in the early stages of the model. Methods DMM surgery was performed in the right knee of 10-week-old male wild-type (WT), Ccr2 null, or Ccr2RFP C57BL/6 mice. Knee hyperalgesia was measured using a Pressure Application Measurement device. CCR2 receptor antagonist (CCR2RA) was injected systemically (i.p.) or intra-articularly (i.a.) at different times after DMM to test its ability to reverse knee hyperalgesia. In vivo Ca2+ imaging of the dorsal root ganglion (DRG) was performed to assess sensory neuron responses to CCL2 injected into the knee joint cavity. CCL2 protein in the knee was measured by ELISA. Ccr2RFP mice and immunohistochemical staining for the pan-neuronal marker, protein gene product 9.5 (PGP9.5), or the sensory neuron marker, calcitonin gene-related peptide (CGRP), were used to visualize the location of CCR2 on intra-articular afferents. Results WT, but not Ccr2 null, mice displayed knee hyperalgesia 2–16 weeks after DMM. CCR2RA administered i.p. alleviated established hyperalgesia in WT mice 4 and 8 weeks after surgery. Intra-articular injection of CCL2 excited sensory neurons in the L4-DRG, as determined by in vivo calcium imaging; responses to CCL2 increased in mice 20 weeks after DMM. CCL2, but not vehicle, injected i.a. rapidly caused transient knee hyperalgesia in naïve WT, but not Ccr2 null, mice. Intra-articular CCR2RA injection also alleviated established hyperalgesia in WT mice 4 and 7 weeks after surgery. CCL2 protein was elevated in the knees of both WT and Ccr2 null mice 4 weeks after surgery. Co-expression of CCR2 and PGP9.5 as well as CCR2 and CGRP was observed in the lateral synovium of naïve mice; co-expression was also observed in the medial compartment of knees 8 weeks after DMM. Conclusions The findings suggest that CCL2-CCR2 signaling locally in the joint contributes to knee hyperalgesia in experimental OA, and it is in part mediated through direct stimulation of CCR2 expressed by intra-articular sensory afferents. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02486-y.
Collapse
Affiliation(s)
- Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA
| | - Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA
| | - David L Wokosin
- Department of Physiology, Northwestern University, Chicago, IL, 60611, USA
| | - Dongjun Ren
- Department of Pharmacology, Northwestern University, Chicago, IL, 60611, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Chicago, IL, 60611, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA.
| |
Collapse
|
37
|
Khella CM, Asgarian R, Horvath JM, Rolauffs B, Hart ML. An Evidence-Based Systematic Review of Human Knee Post-Traumatic Osteoarthritis (PTOA): Timeline of Clinical Presentation and Disease Markers, Comparison of Knee Joint PTOA Models and Early Disease Implications. Int J Mol Sci 2021; 22:1996. [PMID: 33671471 PMCID: PMC7922905 DOI: 10.3390/ijms22041996] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
Understanding the causality of the post-traumatic osteoarthritis (PTOA) disease process of the knee joint is important for diagnosing early disease and developing new and effective preventions or treatments. The aim of this review was to provide detailed clinical data on inflammatory and other biomarkers obtained from patients after acute knee trauma in order to (i) present a timeline of events that occur in the acute, subacute, and chronic post-traumatic phases and in PTOA, and (ii) to identify key factors present in the synovial fluid, serum/plasma and urine, leading to PTOA of the knee in 23-50% of individuals who had acute knee trauma. In this context, we additionally discuss methods of simulating knee trauma and inflammation in in vivo, ex vivo articular cartilage explant and in vitro chondrocyte models, and answer whether these models are representative of the clinical inflammatory stages following knee trauma. Moreover, we compare the pro-inflammatory cytokine concentrations used in such models and demonstrate that, compared to concentrations in the synovial fluid after knee trauma, they are exceedingly high. We then used the Bradford Hill Framework to present evidence that TNF-α and IL-6 cytokines are causal factors, while IL-1β and IL-17 are credible factors in inducing knee PTOA disease progresssion. Lastly, we discuss beneficial infrastructure for future studies to dissect the role of local vs. systemic inflammation in PTOA progression with an emphasis on early disease.
Collapse
Affiliation(s)
| | | | | | | | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (C.M.K.); (R.A.); (J.M.H.); (B.R.)
| |
Collapse
|
38
|
Wiegertjes R, van de Loo FAJ, Blaney Davidson EN. A roadmap to target interleukin-6 in osteoarthritis. Rheumatology (Oxford) 2021; 59:2681-2694. [PMID: 32691066 PMCID: PMC7516110 DOI: 10.1093/rheumatology/keaa248] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/26/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023] Open
Abstract
Joint inflammation is present in the majority of OA patients and pro-inflammatory mediators, such as IL-6, are actively involved in disease progression. Increased levels of IL-6 in serum or synovial fluid from OA patients correlate with disease incidence and severity, with IL-6 playing a pivotal role in the development of cartilage pathology, e.g. via induction of matrix-degrading enzymes. However, IL-6 also increases expression of anti-catabolic factors, suggesting a protective role. Until now, this dual role of IL-6 is incompletely understood and may be caused by differential effects of IL-6 classic vs trans-signalling. Here, we review current evidence regarding the role of IL-6 classic- and trans-signalling in local joint pathology of cartilage, synovium and bone. Furthermore, we discuss targeting of IL-6 in experimental OA models and provide future perspective for OA treatment by evaluating currently available IL-6 targeting strategies.
Collapse
Affiliation(s)
- Renske Wiegertjes
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fons A J van de Loo
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Esmeralda N Blaney Davidson
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
39
|
Barker T, Rogers VE, Henriksen VT, Trawick RH, Momberger NG, Lynn Rasmussen G. Circulating IL-10 is compromised in patients predisposed to developing and in patients with severe knee osteoarthritis. Sci Rep 2021; 11:1812. [PMID: 33469085 PMCID: PMC7815723 DOI: 10.1038/s41598-021-81382-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/06/2021] [Indexed: 01/29/2023] Open
Abstract
The purpose of this investigation was to identify if serum interleukin (IL)-10 and tumor necrosis factor (TNF)-α concentrations and their ratio (IL-10/TNF-α) are altered in subjects predisposed to developing knee osteoarthritis following ligamentous injury and in those with severe knee osteoarthritis. Serum IL-10 and TNF-α concentrations were measured in four groups of subjects (n = 218): (1) reportedly-healthy and non-injured control subjects (CON; n = 92), (2) subjects scheduled to undergo anterior cruciate ligament surgery (ACL; n = 42), (3) non-surgical subjects with knee osteoarthritis (OA; n = 60), and (4) subjects with knee osteoarthritis scheduled to undergo total knee arthroplasty (TKA; n = 24). X-ray images were used to grade the severity of knee osteoarthritis. Serum IL-10 and the serum IL-10/TNF-α ratio were significantly lower while serum TNF-α was not significantly perturbed with severe compared to moderate knee osteoarthritis (i.e., Kellgren-Lawrence grade 4 vs. 3, respectively). Serum IL-10 was significantly lower in the absence of serum TNF-α alterations in the ACL group. We conclude that serum IL-10 concentrations are compromised in subjects predisposed to developing knee osteoarthritis following ligamentous trauma and in subjects with radiographic evidence of severe knee osteoarthritis.
Collapse
Affiliation(s)
- Tyler Barker
- grid.420884.20000 0004 0460 774XPrecision Genomics, Intermountain Healthcare, 383 W. Vine Street, Suite #300, Murray, UT 84107 USA ,grid.223827.e0000 0001 2193 0096Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112 USA
| | - Victoria E. Rogers
- grid.416945.b0000 0004 0442 6615The Orthopedic Specialty Hospital, Murray, UT 84107 USA
| | - Vanessa T. Henriksen
- grid.416945.b0000 0004 0442 6615The Orthopedic Specialty Hospital, Murray, UT 84107 USA
| | - Roy H. Trawick
- grid.416945.b0000 0004 0442 6615The Orthopedic Specialty Hospital, Murray, UT 84107 USA ,The Orthopedic Specialty Clinic, Murray, UT 84107 USA
| | - Nathan G. Momberger
- grid.416945.b0000 0004 0442 6615The Orthopedic Specialty Hospital, Murray, UT 84107 USA ,The Orthopedic Specialty Clinic, Murray, UT 84107 USA
| | - G. Lynn Rasmussen
- grid.416945.b0000 0004 0442 6615The Orthopedic Specialty Hospital, Murray, UT 84107 USA ,The Orthopedic Specialty Clinic, Murray, UT 84107 USA
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Current thinking in the study of posttraumatic osteoarthritis (PTOA) is overviewed: the osteoarthritis which follows acute joint injury. The review particularly highlights important publications in the last 18 months, also reflecting on key older literature, in terms of what have we have we learned and have yet to learn from PTOA, which can advance the osteoarthritis field as a whole. RECENT FINDINGS PTOA is a mechanically driven disease, giving insight into mechanical drivers for osteoarthritis. A mechanosensitive molecular tissue injury response (which includes activation of pain, degradative and also repair pathways) is triggered by acute joint injury and seen in osteoarthritis. Imaging features of PTOA are highly similar to osteoarthritis, arguing against it being a different phenotype. The inflammatory pathways activated by injury contribute to early joint symptoms. However, later structural changes appear to be dissociated from traditional measures of synovial inflammation. SUMMARY PTOA remains an important niche in which to understand processes underlying osteoarthritis and seek interventional targets. Whether PTOA has true molecular or clinical differences to osteoarthritis as a whole remains to be understood. This knowledge is important for a field where animal modelling of the disease relies heavily on the link between injury and osteoarthritis.
Collapse
Affiliation(s)
- Fiona E Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Zhu L, Donhou S, Burleigh A, Miotla Zarebska J, Curtinha M, Parisi I, Khan SN, Dell'Accio F, Chanalaris A, Vincent TL. TSG-6 Is Weakly Chondroprotective in Murine OA but Does not Account for FGF2-Mediated Joint Protection. ACR Open Rheumatol 2020; 2:605-615. [PMID: 33029956 PMCID: PMC7571392 DOI: 10.1002/acr2.11176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/11/2020] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Tumor necrosis factor α-stimulated gene 6 (TSG-6) is an anti-inflammatory protein highly expressed in osteoarthritis (OA), but its influence on the course of OA is unknown. METHODS Cartilage injury was assessed by murine hip avulsion or by recutting rested explants. Forty-two previously validated injury genes were quantified by real-time polymerase chain reaction in whole joints following destabilization of the medial meniscus (DMM) (6 hours and 7 days). Joint pathology was assessed at 8 and 12 weeks following DMM in 10-week-old male and female fibroblast growth factor 2 (FGF2)-/- , TSG-6-/- , TSG-6tg (overexpressing), FGF2-/- ;TSG-6tg (8 weeks only) mice, as well as strain-matched, wild-type controls. In vivo cartilage repair was assessed 8 weeks following focal cartilage injury in TSG-6tg and control mice. FGF2 release following cartilage injury was measured by enzyme-linked immunosorbent assay. RESULTS TSG-6 messenger RNA upregulation was strongly FGF2-dependent upon injury in vitro and in vivo. Fifteeen inflammatory genes were significantly increased in TSG-6-/- joints, including IL1α, Ccl2, and Adamts5 compared with wild type. Six genes were significantly suppressed in TSG-6-/- joints including Timp1, Inhibin βA, and podoplanin (known FGF2 target genes). FGF2 release upon cartilage injury was not influenced by levels of TSG-6. Cartilage degradation was significantly increased at 12 weeks post-DMM in male TSG-6-/- mice, with a nonsignificant 30% reduction in disease seen in TSG-6tg mice. No differences were observed in cartilage repair between genotypes. TSG-6 overexpression was unable to prevent accelerated OA in FGF2-/- mice. CONCLUSION TSG-6 influences early gene regulation in the destabilized joint and exerts a modest late chondroprotective effect. Although strongly FGF2 dependent, TSG-6 does not explain the strong chondroprotective effect of FGF2.
Collapse
Affiliation(s)
- Linyi Zhu
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Shannah Donhou
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Annika Burleigh
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Jadwiga Miotla Zarebska
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Marcia Curtinha
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Ida Parisi
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Sumayya Nafisa Khan
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | | | - Anastasios Chanalaris
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Tonia L Vincent
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| |
Collapse
|
42
|
Yarmola EG, Shah YY, Lakes EH, Pacheco YC, Xie DF, Dobson J, Allen KD. Use of magnetic capture to identify elevated levels of CCL2 following intra-articular injection of monoiodoacetate in rats. Connect Tissue Res 2020; 61:485-497. [PMID: 31438731 PMCID: PMC7036010 DOI: 10.1080/03008207.2019.1620223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/25/2019] [Indexed: 02/03/2023]
Abstract
PURPOSE Synovial fluid biomarkers help evaluate osteoarthritis (OA) development. Magnetic capture, our new magnetic nanoparticle-based technology, has proven to be effective for determining extracellular matrix fragment levels in two rat OA models. Here, the feasibility of magnetic capture for detecting monocyte chemoattractant protein-1 (MCP-1 or CCL2) is demonstrated after intra-articular injection of monoiodoacetate (MIA) in the rat knee. METHODS Forty-eight male Lewis rats received a right hind limb, intra-articular injection of MIA (1 mg in 25 µl of saline) or 25 µl of saline. Magnetic capture and lavage were performed at 7 days after injection (n = 6 per treatment per procedure), with magnetic capture additionally performed at 14 and 28 days post-injection (n = 6 per treatment per time point). CCL2 was also assessed in serum. RESULTS Serum CCL2 levels revealed no difference between MIA and saline animals (p = 0.0851). In contrast, magnetic capture and lavage detected a significant increase of CCL2 in the MIA-injected knee, with the MIA-injected knee having elevated CCL2 compared to contralateral and saline-injected knees (p = 0.00016 (contralateral) and p = 0.00016 (saline) for magnetic capture; p = 0.00023 (contralateral) and p = 0.00049 (saline) for lavage). CONCLUSIONS Magnetic capture of CCL2 was successfully developed and applied to determine levels of CCL2 in a rat knee. Magnetic capture detected a statistically significant increase of CCL2 in MIA-injected knees compared to controls, and CCL2 levels stayed relatively stable from week 1 through week 4 post-MIA injection.
Collapse
Affiliation(s)
- Elena G. Yarmola
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Yash Y. Shah
- Department of Materials Science and Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Emily H. Lakes
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Yan C. Pacheco
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Danny F. Xie
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Jon Dobson
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
- Department of Materials Science and Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Kyle D. Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
43
|
King JD, Rowland G, Villasante Tezanos AG, Warwick J, Kraus VB, Lattermann C, Jacobs CA. Joint Fluid Proteome after Anterior Cruciate Ligament Rupture Reflects an Acute Posttraumatic Inflammatory and Chondrodegenerative State. Cartilage 2020; 11:329-337. [PMID: 30033738 PMCID: PMC7298591 DOI: 10.1177/1947603518790009] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE The purpose of this study was to evaluate changes in the synovial fluid proteome following acute anterior cruciate ligament (ACL) injury. DESIGN This study represents a secondary analysis of synovial fluid samples collected from the placebo group of a previous randomized trial. Arthrocentesis was performed twice on 6 patients with an isolated acute ACL tear at a mean of 6 and 14 days postinjury. Synovial fluid was analyzed by a highly multiplexed assay of 1129 proteins (SOMAscan version 3, SomaLogic, Inc., Boulder, CO). Pathway analysis using DAVID was performed; genes included met 3 criteria: significant change between the 2 study time points using a paired t test, significant change between the 2 study time points using a Mann-Whitney nonparametric test, and significant Benjamini post hoc analysis. RESULTS Fifteen analytes demonstrated significant increases between time points. Five of the 15 have been previously associated with the onset and/or severity of rheumatoid arthritis, including apoliopoprotein E and isoform E3, vascular cell adhesion protein 1, interleukin-34, and cell surface glycoprotein CD200 receptor 1. Chondrodegenerative enzymes and products of cartilage degeneration all increased over time following injury: MMP-1 (P = 0.08, standardized response mean [SRM] = 1.00), MMP-3 (P = 0.05, SRM = 0.90), ADAM12 (P = 0.03, SRM = 1.31), aggrecan (P = 0.08, SRM = 1.13), and CTX-II (P = 0.07, SRM = 0.56). Notable pathways that were differentially expressed following injury were the cytokine-cytokine receptor interaction and osteoclast differentiation pathways. CONCLUSIONS The proteomic results and pathway analysis demonstrated a pattern of cartilage degeneration, not only consistent with previous findings but also changes consistent with an inflammatory arthritogenic process post-ACL injury.
Collapse
Affiliation(s)
- John D. King
- Department of Orthopedic Surgery,
University of Kentucky, Lexington, KY, USA
| | - Grant Rowland
- Central Texas Sports Medicine &
Orthopedics, Bryan, TX, USA
| | | | - James Warwick
- College of Medicine, University of
Kentucky, Lexington, KY, USA
| | - Virginia B. Kraus
- Duke Molecular Physiology Institute,
Department of Medicine, Duke University School of Medicine, Durham, NC, USA,Division of Rheumatology, Department of
Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Christian Lattermann
- Department of Orthopaedic Surgery,
Harvard Medical School and Brigham and Women’s Hosptial, Chestnut Hill, MS,
USA
| | - Cale A. Jacobs
- Department of Orthopedic Surgery,
University of Kentucky, Lexington, KY, USA,Cale A. Jacobs, Department of Orthopedic
Surgery & Sports Medicine, University of Kentucky, 740 South Limestone
Street, Room K426, Lexington, KY 40536-0284, USA.
| |
Collapse
|
44
|
Abdallah FW, Gilron I, Fillingim RB, Tighe P, Parvataneni HK, Ghasemlou N, Sawhney M, McCartney CJL. AAAPT Diagnostic Criteria for Acute Knee Arthroplasty Pain. PAIN MEDICINE (MALDEN, MASS.) 2020; 21:1049-1060. [PMID: 32022891 PMCID: PMC8453639 DOI: 10.1093/pm/pnz355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
OBJECTIVE The relationship between preexisting osteoarthritic pain and subsequent post-total knee arthroplasty (TKA) pain is not well defined. This knowledge gap makes diagnosis of post-TKA pain and development of management plans difficult and may impair future investigations on personalized care. Therefore, a set of diagnostic criteria for identification of acute post-TKA pain would inform standardized management and facilitate future research. METHODS The Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership with the US Food and Drug Administration (FDA), the American Pain Society (APS), and the American Academy of Pain Medicine (AAPM) formed the ACTTION-APS-AAPM Pain Taxonomy (AAAPT) initiative to address this goal. A multidisciplinary work group of pain experts was invited to conceive diagnostic criteria and dimensions of acute post-TKA pain. RESULTS The working group used contemporary literature combined with expert opinion to generate a five-dimensional taxonomical structure based upon the AAAPT framework (i.e., core diagnostic criteria, common features, modulating factors, impact/functional consequences, and putative mechanisms) that characterizes acute post-TKA pain. CONCLUSIONS The diagnostic criteria created are proposed to define the nature of acute pain observed in patients following TKA.
Collapse
Affiliation(s)
- Faraj W Abdallah
- Department of Anesthesiology and Pain Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
| | - Ian Gilron
- Department of Anesthesiology & Perioperative Medicine
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | | | | | - Hari K Parvataneni
- Department of Orthopedic Surgery and Rehabilitation, University of Florida, Gainesville, Florida, USA
| | - Nader Ghasemlou
- Department of Anesthesiology & Perioperative Medicine
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Mona Sawhney
- School of Nursing & Department of Anesthesiology and Perioperative Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Colin J L McCartney
- Department of Anesthesiology and Pain Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
| |
Collapse
|
45
|
Identifying effector molecules, cells, and cytokines of innate immunity in OA. Osteoarthritis Cartilage 2020; 28:532-543. [PMID: 32044352 DOI: 10.1016/j.joca.2020.01.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 02/06/2023]
Abstract
Inflammatory changes are observed in affected joints of osteoarthritis (OA) patients and are thought to be involved in the pathology that develops along OA progression. This narrative review provides an overview of the various cell types that are present in the joint during OA and which alarmins, cytokines, chemokines, growth factors, and other mediators they produce. Moreover, the involvement of more systemic processes like inflammaging and its associated cellular senescence in the context of OA are discussed.
Collapse
|
46
|
Labinsky H, Panipinto PM, Ly KA, Khuat DK, Madarampalli B, Mahajan V, Clabeaux J, MacDonald K, Verdin PJ, Buckner JH, Noss EH. Multiparameter Analysis Identifies Heterogeneity in Knee Osteoarthritis Synovial Responses. Arthritis Rheumatol 2020; 72:598-608. [PMID: 31702112 DOI: 10.1002/art.41161] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 11/05/2019] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Synovial membrane inflammation is common in osteoarthritis (OA) and increases cartilage injury. However, synovial fluid and histology studies suggest that OA inflammatory responses are not homogeneous. Greater understanding of these responses may provide new insights into OA disease mechanisms. We undertook this study to develop a novel multiparameter approach to phenotype synovial responses in knee OA. METHODS Cell composition and soluble protein production were measured by flow cytometry and multiplex enzyme-linked immunosorbent assay in synovium collected from OA patients undergoing knee replacement surgery (n = 35). RESULTS Testing disaggregation conditions showed that aggressive digestion improved synovial cell yield and mesenchymal staining by flow cytometry, but it negatively impacted CD4+ T cell and CD56+ natural killer cell staining. Less aggressive digestion preserved these markers and showed highly variable T cell infiltration (range 0-43%; n = 32). Correlation analysis identified mesenchymal subpopulations associated with different nonmesenchymal populations, including macrophages and T cells (CD45+CD11b+HLA-DR+ myeloid cells with PDPN+CD73+CD90-CD34- mesenchymal cells [r = 0.65, P < 0.0001]; and CD45+CD3+ T cells with PDPN+CD73+CD90+CD34+ mesenchymal cells [r = 0.50, P = 0.003]). Interleukin-6 (IL-6) measured by flow cytometry strongly correlated with IL-6 released by ex vivo culture of synovial tissue (r = 0.59, P = 0.0012) and was highest in mesenchymal cells coexpressing CD90 and CD34. IL-6, IL-8, complement factor D, and IL-10 release correlated positively with tissue cellularity (P = 0.0042, P = 0.018, P = 0.0012, and P = 0.038, respectively). Additionally, increased CD8+ T cell numbers correlated with retinol binding protein 4 (P = 0.033). Finally, combining flow cytometry and multiplex data identified patient clusters with different types of inflammatory responses. CONCLUSION We used a novel approach to analyze OA synovium, identifying patient-specific inflammatory clusters. Our findings indicate that phenotyping synovial inflammation may provide new insights into OA patient heterogeneity and biomarker development.
Collapse
Affiliation(s)
- Hannah Labinsky
- University of Washington, Seattle, and University Medical Center of Johannes Gutenberg University, Germany
| | | | - Kaytlyn A Ly
- Virginia Mason Medical Center, Seattle, Washington
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Struglics A, Turkiewicz A, Larsson S, Lohmander LS, Roemer FW, Frobell R, Englund M. Molecular and imaging biomarkers of local inflammation at 2 years after anterior cruciate ligament injury do not associate with patient reported outcomes at 5 years. Osteoarthritis Cartilage 2020; 28:356-362. [PMID: 31940458 DOI: 10.1016/j.joca.2019.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/19/2019] [Accepted: 12/19/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To estimate the association between molecular or imaging inflammatory biomarkers at 2 years after anterior cruciate ligament (ACL) injury and patient-reported outcomes at 5 years. METHODS For 116 ACL-injured patients, molecular biomarkers of inflammation (synovial fluid and serum cytokines) and Hoffa- and effusion-synovitis as visualized on magnetic resonance imaging (MRI) were assessed 2 years post-injury. Knee injury and Osteoarthritis Outcome Score (KOOS) and SF-36 were assessed at 2 and 5 years. We used multiple imputation to handle biomarker values that were below the level of detection or missing, and linear regression for statistical analyses. RESULTS None of the synovial fluid cytokines or imaging biomarkers of inflammation at 2 years were associated with any of the patient-reported outcomes at 5 years. With each log10 unit higher of serum tumor necrosis factor concentration the knee-related quality of life of KOOS was increased (i.e., better outcome) by 35 (95% confidence interval 7 to 63) points. No other serum biomarker measured at 2 years was associated with patient-reported outcome at 5 years. CONCLUSION Local joint inflammation assessed by biomarkers in synovial fluid and Hoffa- and effusion-synovitis on MRI at 2 years after an ACL injury did not associate with patient-reported outcomes at 5 years. Thus, chronic inflammation in the ACL-injured knee, as reflected by the biomarkers studied here, seems not to be a key determinant for the long-term patient-reported outcomes.
Collapse
Affiliation(s)
- A Struglics
- Orthopaedics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| | - A Turkiewicz
- Clinical Epidemiology Unit, Orthopaedics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - S Larsson
- Orthopaedics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - L S Lohmander
- Orthopaedics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - F W Roemer
- Orthopaedics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden; Quantitative Imaging Center (QIC), Department of Radiology, Boston University School of Medicine, Boston, MA, USA; Department of Radiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - R Frobell
- Orthopaedics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - M Englund
- Clinical Epidemiology Unit, Orthopaedics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
48
|
Watt FE, Hamid B, Garriga C, Judge A, Hrusecka R, Custers RJH, Jansen MP, Lafeber FP, Mastbergen SC, Vincent TL. The molecular profile of synovial fluid changes upon joint distraction and is associated with clinical response in knee osteoarthritis. Osteoarthritis Cartilage 2020; 28:324-333. [PMID: 31904489 PMCID: PMC7054834 DOI: 10.1016/j.joca.2019.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 12/14/2019] [Accepted: 12/22/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Surgical knee joint distraction (KJD) leads to clinical improvement in knee osteoarthritis (OA) and also apparent cartilage regeneration by magnetic resonance imaging. We investigated if alteration of the joint's mechanical environment during the 6 week period of KJD was associated with a molecular response in synovial fluid, and if any change was associated with clinical response. METHOD 20 individuals undergoing KJD for symptomatic radiographic knee OA had SF sampled at baseline, midpoint and endpoint of distraction (6 weeks). SF supernatants were measured by immunoassay for 10 predefined mechanosensitive molecules identified in our previous pre-clinical studies. The composite Knee injury and OA Outcome Score-4 (KOOS4) was collected at baseline, 3, 6 and 12 months. RESULTS 13/20 (65%) were male with mean age 54°±°5yrs. All had Kellgren-Lawrence grade ≥2 knee OA. 6/10 analytes showed statistically significant change in SF over the 6 weeks distraction (activin A; TGFβ-1; MCP-1; IL-6; FGF-2; LTBP2), P < 0.05. Of these, all but activin A increased. Those achieving the minimum clinically important difference of 10 points for KOOS4 over 6 months showed greater increases in FGF-2 and TGFβ-1 than non-responders. An increase in IL-8 during the 6 weeks of KJD was associated with significantly greater improvement in KOOS4 over 12 months. CONCLUSION Detectable, significant molecular changes are observed in SF following KJD, that are remarkably consistent between individuals. Preliminary findings appear to suggest that increases in some molecules are associated with clinically meaningful responses. Joint distraction may provide a potential opportunity in the future to define regenerative biomarker(s) and identify pathways that drive intrinsic cartilage repair.
Collapse
Affiliation(s)
- F E Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, Roosevelt Drive, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, UK.
| | - B Hamid
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, UK.
| | - C Garriga
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, UK.
| | - A Judge
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, UK; Musculoskeletal Research Unit, University of Bristol, UK; National Institute for Health Research Bristol Biomedical Research Centre (NIHR Bristol BRC), University Hospitals Bristol NHS Foundation Trust, UK; MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton, UK.
| | - R Hrusecka
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, UK.
| | - R J H Custers
- Department of Orthopaedic Surgery, University Medical Center Utrecht, the Netherlands.
| | - M P Jansen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, the Netherlands.
| | - F P Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, the Netherlands.
| | - S C Mastbergen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, the Netherlands.
| | - T L Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, UK.
| |
Collapse
|
49
|
Pathomechanisms of Posttraumatic Osteoarthritis: Chondrocyte Behavior and Fate in a Precarious Environment. Int J Mol Sci 2020; 21:ijms21051560. [PMID: 32106481 PMCID: PMC7084733 DOI: 10.3390/ijms21051560] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
Traumatic injuries of the knee joint result in a wide variety of pathomechanisms, which contribute to the development of so-called posttraumatic osteoarthritis (PTOA). These pathogenetic processes include oxidative stress, excessive expression of catabolic enzymes, release of damage-associated molecular patterns (DAMPs), and synovial inflammation. The present review focuses on the underlying pathomechanisms of PTOA and in particular the behavior and fate of the surviving chondrocytes, comprising chondrocyte metabolism, regulated cell death, and phenotypical changes comprising hypertrophy and senescence. Moreover, possible therapeutic strategies, such as chondroanabolic stimulation, anti-oxidative and anti-inflammatory treatment, as well as novel therapeutic targets are discussed.
Collapse
|
50
|
Snoeker B, Turkiewicz A, Magnusson K, Frobell R, Yu D, Peat G, Englund M. Risk of knee osteoarthritis after different types of knee injuries in young adults: a population-based cohort study. Br J Sports Med 2019; 54:725-730. [DOI: 10.1136/bjsports-2019-100959] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2019] [Indexed: 01/16/2023]
Abstract
ObjectivesTo estimate the risk of clinically diagnosed knee osteoarthritis (OA) after different types of knee injuries in young adults.MethodsIn a longitudinal cohort study based on population-based healthcare data from Skåne, Sweden, we included all persons aged 25–34 years in 1998–2007 (n=149 288) with and without diagnoses of knee injuries according to International Classification of Diseases (ICD)-10. We estimated the HR of future diagnosed knee OA in injured and uninjured persons using Cox regression, adjusted for potential confounders. We also explored the impact of type of injury (contusion, fracture, dislocation, meniscal tear, cartilage tear/other injury, collateral ligament tear, cruciate ligament tear and injury to multiple structures) on diagnosed knee OA risk.ResultsWe identified 5247 persons (mean (SD) age 29.4 (2.9) years, 67% men) with a knee injury and 142 825 persons (mean (SD) age 30.2 (3.0) years, 45% men) without. We found an adjusted HR of 5.7 (95% CI 5.0 to 6.6) for diagnosed knee OA in injured compared with uninjured persons during the first 11 years of follow-up and 3.4 (95% CI 2.9 to 4.0) during the following 8 years. The corresponding risk difference (RD) after 19 years of follow-up was 8.1% (95% CI 6.7% to 9.4%). Cruciate ligament injury, meniscal tear and fracture of the tibia plateau/patella were associated with greatest increase in risk (RD of 19.6% (95% CI 13.2% to 25.9%), 10.5% (95% CI 6.4% to 14.7%) and 6.6% (95% CI 1.1% to 12.2%), respectively).ConclusionIn young adults, knee injury increases the risk of future diagnosed knee OA about sixfold with highest risks found after cruciate ligament injury, meniscal tear and intra-articular fracture.
Collapse
|