1
|
Guo H, Wu Z, Shen G, Huang Z, Zha Z, Zheng X, Luo X, Mai B, Ye J, Li J. Perfluoroalkyl and polyfluoroalkyl substances crossing the blood-joint barrier: Their occurrence and distribution in synovial fluid. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138189. [PMID: 40209403 DOI: 10.1016/j.jhazmat.2025.138189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/14/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Per- and polyfluoroalkyl substances (PFASs) have garnered considerable research attention due to their potential adverse effects on human health. Epidemiological studies have indicated a possible association between PFASs exposure and the prevalence of osteoarthritis (OA). However, the presence of PFASs in the synovial fluid of OA patients and the distribution of PFASs across the blood-joint barrier remains unreported. This study identified significant differences in PFASs profiles between patients and controls, with a markedly higher PFOS-to-PFOA ratio observed in patients. Additionally, the blood-joint transfer efficiency of PFOS was significantly greater in patients than in controls (0.75 vs. 0.53, p < 0.05). Furthermore, PFOS levels were elevated in patients with advanced OA compared to those in the early stages. Positive correlations were observed between synovial fluid PFOS and inflammatory markers C-reactive protein (CRP) and tumor necrosis factor-α (TNF-α), implying that inflammation may facilitate the distribution of PFOS across the joint barrier. This study represents the first documented evidence of the human joint exposure to PFASs and their blood-joint transfer abilities.
Collapse
Affiliation(s)
- Huiying Guo
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China; Guangdong Key Laboratory of Environmental Pollution and Health, College of Environment and Climate, Jinan University, Guangzhou, China
| | - Zifeng Wu
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Guiwang Shen
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zhiyu Huang
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zhengang Zha
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xiaofei Zheng
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xiaojun Luo
- State Key Laboratory of Organic Geochemistry and Guangdong Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, China
| | - Bixian Mai
- State Key Laboratory of Organic Geochemistry and Guangdong Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, China
| | - Jinshao Ye
- Guangdong Key Laboratory of Environmental Pollution and Health, College of Environment and Climate, Jinan University, Guangzhou, China.
| | - Jieruo Li
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
2
|
Bond B, Brown MN, Kurtz MA, Robinson A, Mihalko MJ, Crockarell JR, Holland CT, Guyton JL, Mihalko WM. Osteoarthritic Human Synovial Fluid Alters CoCrMo Electrochemical Properties on a Patient-Specific Basis. J Orthop Res 2025; 43:1144-1154. [PMID: 40195073 DOI: 10.1002/jor.26079] [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: 11/25/2024] [Revised: 02/28/2025] [Accepted: 03/15/2025] [Indexed: 04/09/2025]
Abstract
Cobalt-chromium-molybdenum (CoCrMo) femoral components are widely used in total knee arthroplasty (TKA). However, recent retrospective clinical trials associate moderate adverse local tissue reactions with CoCrMo release in the knee. Additionally, gaps persist in our understanding of the fundamental corrosion processes that occur at the CoCrMo-synovial fluid interface. In this study, we investigated the electrochemical behavior of CoCrMo in human synovial fluid obtained at the time of primary TKA, using CoCrMo in phosphate-buffered saline (PBS) as a comparison. Synovial fluid was collected from 118 patients immediately before arthroplasty, then transferred to a three-electrode electrochemical cell with a wrought CoCrMo alloy working electrode. To quantify electrochemical properties, open circuit potential (OCP), electrochemical impedance spectroscopy, and linear polarization tests were run. Generally, the properties varied on a patient-by-patient basis and significantly differed (p < 0.05) from comparison tests performed in PBS. In human synovial fluid, we measured OCPs between a range of -0.38 and 0.15 V and corrosion potentials (Ecorr) between -0.95 and -0.2 V. Additionally, we reported instantaneous corrosion rates (1/Rp) spanning nearly four orders of magnitude. The variability we documented suggested that the electrochemical properties of CoCrMo implants may depend on the patient's local physiological environment, influenced by the biological and chemical components of synovial fluid. Statement of Clinical Significance: This study shows that human synovial fluid affects the electrochemical behavior of CoCrMo, evidenced by the variation on a patient-by-patient basis. This study may have implications for long-term biological response to orthopaedic implants, including total knee arthroplasty.
Collapse
Affiliation(s)
- Bailey Bond
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee, USA
| | - Madison N Brown
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee, USA
| | - Michael A Kurtz
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Alayna Robinson
- College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Marc J Mihalko
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee, USA
| | - John R Crockarell
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee, USA
| | - Christopher T Holland
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee, USA
| | - James L Guyton
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee, USA
| | - William M Mihalko
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee, USA
| |
Collapse
|
3
|
Bowers R, Berrigan W, Miranda-Comas GE, Jimenez C, Soo Hoo J. Corticosteroid versus ketorolac injection for treatment of knee osteoarthritis flare. PM R 2025. [PMID: 40434241 DOI: 10.1002/pmrj.13387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/03/2025] [Indexed: 05/29/2025]
Affiliation(s)
- Robert Bowers
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - William Berrigan
- Department of Orthopedics, University of California, San Francisco, San Francisco, California, USA
| | - Gerardo E Miranda-Comas
- Department of Physical Medicine, Rehabilitation, and Sports Health, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claudia Jimenez
- Department of Physical Medicine, Rehabilitation, and Sports Health, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Jennifer Soo Hoo
- Department of Rehabilitation Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
4
|
Pan Y, Jin X, Zhou Q, Jin M. The causal relationship between emotions and osteoarthritis: A bidirectional Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42631. [PMID: 40419874 DOI: 10.1097/md.0000000000042631] [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] [Indexed: 05/28/2025] Open
Abstract
Previous study has observed the clinical relationship between affective disorders such as depression and anxiety and osteoarthritis (OA), however the gene causal effect was still unclear. The objective of this study was to evaluate the genetic causal effect of emotions on OA using bidirectional Mendelian randomization (MR), which is a novel methodology elucidating the causal relationship between diseases and screening for the potential driver genes. A bidirectional MR study was conducted to assess the causal effect of emotion on OA. The instrumental variables were selected from publicly available Genome-Wide Association Study summary datasets based on a P-value threshold (P < 5e-8) and linkage disequilibrium clumping criteria (r² < 0.001, window size = 10,000 kb). The robustness of the findings across different MR methods was validated by Cochran Q test, MR-Egger intercept test, MR_PRESSO, F-statistic, and statistical power. The inverse-variance weighted method was employed as the primary analysis due to its weighting approach, which minimizes variance and yields the most precise estimation of causal effects. While MR-Egger (assessed and accounted Pleiotropy), Weighted Median (adjusted for the potential confounding pleiotropy of instrumental variables), Weighted and Simple Mode (identified the effect clusters) methods were used as supplementary analyses to complement the outcome. The main genetic data source consisted of 10,083 participants obtained from publicly accessible repository. The emotions of depression, anxiousness, and hurt were found to have a genetic influence on the development of OA. Specifically, anxiousness was associated with a reduced risk of OA (odds ratio [OR] = 0.486, 95% confidence interval [CI] = 0.260-0.908, P = .024). On the other hand, depression (OR = 2.157, 95% CI = 1.605-2.899, P = 3.4e-07) and hurt (OR = 1.731, 95% CI = 1.092-2.745, P = .020) were identified as genetic factors that increased the susceptibility to OA. The statistical power of depression, anxious and feeling hurt on OA were > 0.99, 0.29, and > 0.99. These findings suggest that genetic factors underlying emotions, particularly depression and emotional distress, significantly influence susceptibility to OA, underscoring the potential for targeted mental health interventions in OA management.
Collapse
Affiliation(s)
- Yu Pan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xiaoliang Jin
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qiujun Zhou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Minwei Jin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Chlorogiannis DD, Taslakian B, Ahmed O, Mukundan A, Guermazi A, Ghodadra A, Krishnasamy VP, Badar W, Epelboym Y. Key Osteoarthritis Biomarkers: What Interventional Radiologists Performing Genicular Artery Embolization Need to Know. Cardiovasc Intervent Radiol 2025:10.1007/s00270-025-04045-3. [PMID: 40399701 DOI: 10.1007/s00270-025-04045-3] [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: 12/09/2024] [Accepted: 04/05/2025] [Indexed: 05/23/2025]
Abstract
Knee osteoarthritis (OA) is a major cause of pain and disability. Genicular artery embolization (GAE) is a promising alternative for patients unresponsive to conservative treatment or unsuitable for total knee replacement. GAE may alleviate knee pain by reducing synovitis and by causing ischemia to pathologic knee nerves. However, outcomes vary based on baseline MRI and radiographic findings, making patient selection challenging. Recently, reduction in synovitis on contrast-enhanced MRI has been associated with pain relief post-GAE. Novel OA phenotypes (inflammatory, cartilage, bone, and atrophic) can aid in patient selection by using imaging and molecular biomarkers. Moreover, imaging features such as cartilage defects, bone marrow lesions, and synovitis may predict symptomatic outcomes. This review explores OA pathophysiology, focusing on radiographic, biochemical, and imaging biomarkers in evaluating GAE for knee OA treatment.
Collapse
Affiliation(s)
- David-Dimitris Chlorogiannis
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 St Francis Str, Boston, MA, 02215, USA.
| | - Bedros Taslakian
- Department of Radiology, Division of Vascular and Interventional Radiology, NYU Langone Health, New York, NY, 10016, USA
| | - Osman Ahmed
- Department of Radiology, University of Chicago Medical Center, University of Chicago, Chicago, IL, 60637, USA
| | - Attur Mukundan
- Division of Rheumatology, Department of Medicine, NYU Langone Health, New York, NY, 10016, USA
| | - Ali Guermazi
- Department of Radiology, VA Boston Healthcare System, Chobanian & Avedisian Boston University School of Medicine, Boston, MA, 02215, USA
| | - Anish Ghodadra
- Department of Radiology, University of Pittsburgh Medical Center, 200 Lothrop Street, Suite E204, Pittsburgh, PA, 15213, USA
| | - Venkatesh P Krishnasamy
- Division of Interventional Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wali Badar
- Department of Radiology, University of Chicago Medical Center, University of Chicago, Chicago, IL, 60637, USA
| | - Yan Epelboym
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 St Francis Str, Boston, MA, 02215, USA
| |
Collapse
|
6
|
Zhao Y, Liu Q, Zhao J, Song D. The roles of natural killer cells in bone and arthritic disease: a narrative review. Immunol Med 2025:1-14. [PMID: 40382682 DOI: 10.1080/25785826.2025.2506260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/28/2025] [Indexed: 05/20/2025] Open
Abstract
The skeletal system is responsible for the body's support and motor functions, and can be pathologically affected by factors, such as metabolism, autoimmune inflammation, tumors, and infections. Regarding tissue localization and biological function, the immune system is deeply involved in the physiological and pathological processes of the skeletal system. As a regulator and effector cell of the innate immune system, natural killer (NK) cells can exert cytotoxic effects through cell contact and immunomodulatory effects through cytokine secretion. In the past 30 years, many advances have been made regarding the role of NK cells and their derived cytokines on bone and joints. In this review, the role of NK cells in the physiological activities of bone remodeling is summarized first, focusing on osteoclast differentiation and function. Subsequently, the roles of NK cells in osteoarthritis, bone tumors, and bone diseases caused by microbial infections are described, meanwhile, some conflicting research results are discussed. By reviewing the state-of-the-art progress of NK cells in the above-mentioned bone physiological and pathological processes, it is helpful to clarify the blind spots of current research and provide some references for the integrated evaluation of immune factors in the study of skeletal system diseases.
Collapse
Affiliation(s)
- Yiming Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P. R. China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, P. R. China
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P. R. China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, P. R. China
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P. R. China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, P. R. China
| | - Dezhi Song
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P. R. China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, P. R. China
| |
Collapse
|
7
|
Gems D. How aging causes osteoarthritis: An evolutionary physiology perspective. Osteoarthritis Cartilage 2025:S1063-4584(25)01024-6. [PMID: 40381687 DOI: 10.1016/j.joca.2025.05.001] [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: 01/20/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
Late-life diseases result from the poorly understood process of senescence (aging), which is largely genetically determined. According to a recently proposed evolutionary physiology-based account, the multifactorial model, senescence is largely caused by evolved but non-adaptive programmatic mechanisms specified by the wild-type (i.e. normal) genome. These act together with disruptions to wild-type function (due e.g. to infectious pathogens, mechanical injury and malnutrition) in a variety of combinations to generate diverse late-life diseases. Here, I explore the utility of this model by testing its capacity to provide an account of one complex, late-life disease, osteoarthritis (OA), and suggest a framework for understanding OA etiology. In this cartilage-focused framework, a core OA disease mechanism is a futile (non-adaptive) developmental program of endochondral ossification, in which hypertrophic articular cartilage chondrocytes alter joint architecture. Programmatic changes prime chondrocytes for futile program activation, which can be triggered by secondary causes of OA (e.g. joint mechanical injury). I suggest that an evolutionary cause of this priming, involving antagonistic pleiotropy, is selection to maximize early-life tissue repair benefits at the expense of late-life programmatic costs.
Collapse
Affiliation(s)
- David Gems
- Institute of Healthy Ageing, and Department of Genetics, Evolution and Environment, University College London, Gower Street, London, UK.
| |
Collapse
|
8
|
Lin Y, Wu H, Wang J, He W, Hou J, Martin VT, Zhu C, Chen Y, Zhong J, Yu B, Lu A, Guan D, Qin G, Chen W. Nicotinamide Adenine Dinucleotide-Loaded Lubricated Hydrogel Microspheres with a Three-Pronged Approach Alleviate Age-Related Osteoarthritis. ACS NANO 2025; 19:17606-17626. [PMID: 40315404 PMCID: PMC12080321 DOI: 10.1021/acsnano.5c01184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/04/2025]
Abstract
Chondrocyte senescence, synovitis, and decreased level of lubrication play pivotal roles in the pathogenesis of age-related osteoarthritis (AROA). However, there are currently no effective therapeutic interventions capable of altering the progression of OA until it reaches advanced stages, necessitating joint replacement. In this study, lubricious and drug-loaded hydrogel microspheres were designed and fabricated by utilizing microfluidic technology for radical polymerization of chondroitin sulfate methacrylate and incorporating nicotinamide adenine dinucleotide (NAD)-loaded liposomes modified with lactoferrin that are positively charged. Mechanical, tribological, and drug release analyses demonstrated enhanced lubrication properties and an extended drug dissemination time for the NAD@NPs@HM microspheres. In vitro assays unveiled the ability of NAD@NPs@HM to counteract chondrocyte senescence. RNA sequencing analysis, untargeted metabolomics analysis, and in vitro experiments on macrophages revealed that NAD@NPs@HM can regulate the metabolic reprogramming of synovial macrophages, promoting their repolarization from the M1 to M2 phenotype, thereby alleviating synovitis. Intra-articular injection of NAD@NPs@HM in aged mice reduced the mechanisms associated with AROA. These results suggest that NAD@NPs@HM may provide extended drug release, improved joint lubrication leading to better gait, and attenuation of AROA pathogenic processes, indicating its potential as a therapeutic approach for AROA.
Collapse
Affiliation(s)
- Yanpeng Lin
- Department
of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Hangtian Wu
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Jun Wang
- School
of Animal Science and Technology, Foshan
University, Foshan, Guangdong 528231, People’s Republic of China
| | - Wanling He
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Guangdong
Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jiahui Hou
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Vidmi Taolam Martin
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Chencheng Zhu
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Yupeng Chen
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Guangdong
Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, P. R. China
| | - Junyuan Zhong
- Department
of Medical Imaging, Ganzhou People’s
Hospital, Ganzhou, Jiangxi 341000, P. R. China
| | - Bin Yu
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Aiping Lu
- Institute
of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong 999077, P. R. China
- Guangdong-Hong
Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510515, P. R. China
| | - Daogang Guan
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Guangdong
Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, P. R. China
| | - Genggeng Qin
- Department
of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Weiguo Chen
- Department
of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou, Guangdong 510515, P. R. China
| |
Collapse
|
9
|
Wang Y, Barmin R, Mottaghy FM, Kiessling F, Lammers T, Pallares RM. Nanoparticles in nuclear medicine: From diagnostics to therapeutics. J Control Release 2025; 383:113815. [PMID: 40319914 DOI: 10.1016/j.jconrel.2025.113815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/13/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
The use of nanoparticles in nuclear medicine is paradoxical. While several nanoformulations, such as 99mTc colloids, have been used for diagnosis for decades, only a few new radionanomedicines have been able to reach the market, despite extensive preclinical efforts. This contradiction is dictated by the unique features of nanoparticles, such as (potential) prolonged circulation times, slow compartment exchanges, and large accumulations in the mononuclear phagocyte system, which allow for certain specific applications while preventing others. In this review, we discuss the development and clinical application of radiolabeled nanoparticles as imaging agents for disease diagnosis and patient stratification, as well as their promise and potential to be used as next-generation formulations to improve the efficacy of radiotherapy.
Collapse
Affiliation(s)
- Ying Wang
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Roman Barmin
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen 52074, Germany; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Roger M Pallares
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany.
| |
Collapse
|
10
|
Li W, Liu Y, Wei M, Yang Z, Li Z, Guo Z, Yan L, Lu Y, Tang H, Li B, Huang W. Functionalized Biomimetic Nanoparticles Targeting the IL-10/IL-10Rα/Glycolytic Axis in Synovial Macrophages Alleviate Cartilage Degeneration in Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504768. [PMID: 40317692 DOI: 10.1002/advs.202504768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Indexed: 05/07/2025]
Abstract
Osteoarthritis (OA) is a low-grade inflammatory disease that is highly associated with severe hyperplasia of the synovial membrane and the degeneration of cartilage. Interleukin-10 (IL-10), has been extensively studied, while its receptor, IL-10Rα, has not been widely mentioned in the context of OA. A significant difference is found in the expression of IL-10Rα in synovial macrophages from normal and OA patients, along with a marked increase in the glycolytic activity of synovial macrophages. In IL-10RαLysm OA mice, the specific deficiency of IL-10Rα exacerbated the progression of OA. Mechanistically, hypoxia-inducible factor-1α (HIF-1α) is identified as a key transcription factor, and its inhibition significantly weakened the glycolytic process. Additionally, differences in ferroptosis of chondrocytes are observed. After co-culturing the two types of cells in vitro, a significant connection is found between the glycolytic state of synovial macrophages and the ferroptosis of chondrocytes. To achieve targeted therapy, MI@UN, a biomimetic nanoparticle encapsulating NO-prednisolone in UIO-66-NH2, surface-modified with IL-10, and coated with macrophage membranes (MM), is developed. It significantly slows osteoarthritis progression in mice. This offers new insights into OA pathogenesis, highlighting IL-10Rα as a therapeutic target and supporting MI@UN's translational use for OA treatment.
Collapse
Affiliation(s)
- Wenwei Li
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
| | - Yang Liu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Ming Wei
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Zhichao Yang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Zhaoyu Li
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Zezhong Guo
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
| | - Liang Yan
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yang Lu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Hao Tang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Bofeng Li
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, 230001, China
| | - Wei Huang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| |
Collapse
|
11
|
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z, Li N. Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res 2025; 71:227-262. [PMID: 38876191 DOI: 10.1016/j.jare.2024.06.010] [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: 02/17/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND As people age, degenerative bone and joint diseases (DBJDs) become more prevalent. When middle-aged and elderly people are diagnosed with one or more disorders such as osteoporosis (OP), osteoarthritis (OA), and intervertebral disc degeneration (IVDD), it often signals the onset of prolonged pain and reduced functionality. Chronic inflammation has been identified as the underlying cause of various degenerative diseases, including DBJDs. Recently, excessive activation of pyroptosis, a form of programed cell death (PCD) mediated by inflammasomes, has emerged as a primary driver of harmful chronic inflammation. Consequently, pyroptosis has become a potential target for preventing and treating DBJDs. AIM OF REVIEW This review explored the physiological and pathological roles of the pyroptosis pathway in bone and joint development and its relation to DBJDs. Meanwhile, it elaborated the molecular mechanisms of pyroptosis within individual cell types in the bone marrow and joints, as well as the interplay among different cell types in the context of DBJDs. Furthermore, this review presented the latest compelling evidence supporting the idea of regulating the pyroptosis pathway for DBJDs treatment, and discussed the potential, limitations, and challenges of various therapeutic strategies involving pyroptosis regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW In summary, an interesting identity for the unregulated pyroptosis pathway in the context of DBJDs was proposed in this review, which was undertaken as a spoiler of peaceful coexistence between cells in a degenerative environment. Over the extended course of DBJDs, pyroptosis pathway perpetuated its activity through crosstalk among pyroptosis cascades in different cell types, thus exacerbating the inflammatory environment throughout the entire bone marrow and joint degeneration environment. Correspondingly, pyroptosis regulation therapy emerged as a promising option for clinical treatment of DBJDs.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jilin Fan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000 China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300 China.
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
12
|
Gao Q, Zhang X, Makarcyzk MJ, Wong LE, Quig MSV, Shinohara I, Murayama M, Chow SKH, Bunnell BA, Lin H, Goodman SB. Macrophage phenotypes modulate neoangiogenesis and fibroblast profiles in synovial-like organoid cultures. Osteoarthritis Cartilage 2025; 33:590-600. [PMID: 39978574 DOI: 10.1016/j.joca.2025.02.777] [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: 07/12/2024] [Revised: 01/28/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVE To investigate the interaction among the cells thought to be foundational to inflammation, fibrosis, and angiogenesis in the synovial membrane. METHOD We encapsulated fibroblasts, polarized macrophages, and endothelial cells in a 3D culture system. We used this model to determine the cellular transcriptional profiles, cytokine secretion, and vascular formation associated with different macrophage phenotype conditions. RESULTS Neo-angiogenesis reached its maximum level at approximately day 21 in the presence of pro-inflammatory macrophages conditions, but was sustained in the presence of anti-inflammatory macrophages. RNA sequencing revealed an influence of macrophage phenotype on gene expression associated with fibrosis and angiogenesis. Furthermore, by including lipopolysaccharides-coated polyethylene particles (lcPE), an inflammatory stimulus replicating wear debris from joint replacements into our system, insights into the local reaction to byproducts of different biomaterials can be ascertained. CONCLUSION Chronic inflammation and fibrosis of the synovial membrane are often present in osteoarthritis and post-total joint arthroplasty. Our results suggest that the progression of inflammatory synovial diseases is influenced by macrophage phenotypes.
Collapse
Affiliation(s)
- Qi Gao
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA.
| | - Xiurui Zhang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| | - Meagan J Makarcyzk
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| | - Laurel Elizabeth Wong
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA.
| | - Madison Sidney Virgil Quig
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA; Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| | - Issei Shinohara
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA.
| | - Masatoshi Murayama
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA.
| | - Simon Kwoon-Ho Chow
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA.
| | - Bruce A Bunnell
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| | - Stuart B Goodman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA.
| |
Collapse
|
13
|
Sourugeon Y, Boffa A, Perucca Orfei C, de Girolamo L, Magalon J, Sánchez M, Tischer T, Filardo G, Laver L. Cell-based therapies have disease-modifying effects on osteoarthritis in animal models: A systematic review by the ESSKA Orthobiologic Initiative. Part 3: Umbilical cord, placenta, and other sources for cell-based injectable therapies. Knee Surg Sports Traumatol Arthrosc 2025; 33:1695-1708. [PMID: 39302089 PMCID: PMC12022835 DOI: 10.1002/ksa.12472] [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: 07/15/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE This systematic review aimed to investigate in animal models the presence of disease-modifying effects driven by non-bone marrow-derived and non-adipose-derived products, with a particular focus on umbilical cord and placenta-derived cell-based therapies for the intra-articular injective treatment of osteoarthritis (OA). METHODS A systematic review was performed on three electronic databases (PubMed, Web of Science and Embase) according to PRISMA guidelines. The results were synthesised to investigate disease-modifying effects in preclinical animal studies comparing injectable umbilical cord, placenta, and other sources-derived products with OA controls. The risk of bias was assessed using the SYRCLE tool. RESULTS A total of 80 studies were included (2314 animals). Cell therapies were most commonly obtained from the umbilical cord in 33 studies and placenta/amniotic tissue in 18. Cell products were xenogeneic in 61 studies and allogeneic in the remaining 19 studies. Overall, 25/27 (92.6%) of studies on umbilical cord-derived products documented better results compared to OA controls in at least one of the following outcomes: macroscopic, histological and/or immunohistochemical findings, with 19/22 of studies (83.4%) show positive results at the cartilage level and 4/6 of studies (66.7%) at the synovial level. Placenta-derived injectable products documented positive results in 13/16 (81.3%) of the studies, 12/15 (80.0%) at the cartilage level, and 2/4 (50.0%) at the synovial level, but 2/16 studies (12.5%) found overall worse results than OA controls. Other sources (embryonic, synovial, peripheral blood, dental pulp, cartilage, meniscus and muscle-derived products) were investigated in fewer preclinical studies. The risk of bias was low in 42% of items, unclear in 49%, and high in 9% of items. CONCLUSION Interest in cell-based injectable therapies for OA treatment is soaring, particularly for alternatives to bone marrow and adipose tissue. While expanded umbilical cord mesenchymal stem cells reported auspicious disease-modifying effects in preventing OA progression in animal models, placenta/amniotic tissue also reported deleterious effects on OA joints. Lower evidence has been found for other cellular sources such as embryonic, synovial, peripheral blood, dental-pulp, cartilage, meniscus, and muscle-derived products. LEVEL OF EVIDENCE Level II.
Collapse
Affiliation(s)
- Yosef Sourugeon
- Division of Surgery, Orthopaedics DepartmentChaim Sheba Medical CentreRamat GanIsrael
| | - Angelo Boffa
- Applied and Translational Research Center, IRCCS Istituto Ortopedico RizzoliBolognaItaly
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Carlotta Perucca Orfei
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Ospedale Galeazzi Sant'AmbrogioMilanItaly
| | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Ospedale Galeazzi Sant'AmbrogioMilanItaly
| | - Jeremy Magalon
- INSERM, NRA, C2VNAix Marseille UniversityMarseilleFrance
- SAS RemedexMarseilleFrance
- Cell Therapy Laboratory, Hôpital De La Conception, AP‐HMMarseilleFrance
| | - Mikel Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas VitoriaVitoria‑GasteizSpain
- Arthroscopic Surgery Unit, Hospital Vithas VitoriaVitoria‐GasteizSpain
| | - Thomas Tischer
- Department of Orthopaedic and Trauma SurgeryMalteser WaldkrankenhausErlangenGermany
- Department of Orthopaedic SurgeryUniversity of RostockRostockGermany
| | - Giuseppe Filardo
- Applied and Translational Research Center, IRCCS Istituto Ortopedico RizzoliBolognaItaly
- Department of SurgeryService of Orthopaedics and Traumatology, EOCLuganoSwitzerland
- Faculty of Biomedical SciencesUniversità Della Svizzera ItalianaLuganoSwitzerland
| | - Lior Laver
- Arthrosport ClinicTel‑AvivIsrael
- Rappaport Faculty of Medicine, Technion University Hospital (IsraelInstitute of Technology)HaifaIsrael
- Department of OrthopaedicsHillel Yaffe Medical Center (HYMC)HaderaIsrael
| |
Collapse
|
14
|
Usimaki A, Ciamillo SA, Barot D, Linardi RL, Engiles JB, Ortved KF. Single injection of intra-articular autologous protein solution in horses with acute interleukin-1B-induced synovitis decreases joint pathology scores. Equine Vet J 2025; 57:806-816. [PMID: 39051479 PMCID: PMC11982420 DOI: 10.1111/evj.14203] [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: 03/13/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Autologous protein solution (APS) has been shown to decrease lameness in horses with osteoarthritis (OA). Synovitis is an early driver of OA, providing an opportunity to intervene in the progression of disease via intra-articular (IA) therapeutics. OBJECTIVES The objective of this study was to investigate the effects of a single IA APS injection in horses with interleukin-1β (IL-1β)-induced synovitis. We hypothesised that APS would decrease joint swelling and lameness, improve synovial fluid parameters and improve joint pathology scores in horses compared with untreated controls. STUDY DESIGN Randomised controlled in vivo experiment. METHODS Synovitis was induced with IL-1β (65 ng) in one randomly selected tarsocrural joint. Twenty-four hours later, joints were treated with APS (Pro-Stride®) (n = 12) or left as untreated controls (n = 6). Lameness examinations and joint circumference measurements were performed on Days 0 (prior to IL-1β), 1 (prior to APS), 2, 4, 7 and 14. Synovial fluid, obtained on the same days, was analysed for protein concentration, nucleated cell count, and cytokine (IL-1β, TNF-α, IFN-γ, IL-6, IL-10) and prostaglandin E2 (PGE2) concentrations. Gross pathology and synovial membrane histopathology scoring was performed on APS-treated (n = 5), untreated control (n = 4) and normal (n = 9) tarsocrural joints. RESULTS APS did not decrease lameness or joint circumference compared with untreated controls. Synovial fluid parameters were not different between treatment groups. APS treatment did significantly decrease gross and histopathology scores. MAIN LIMITATIONS Main limitations included the use of an induced model of the synovitis, inter-horse variability in the response to IL-1β and likely variability in the constituents of APS from individual horses. CONCLUSIONS APS treatment of tarsocrural joints with synovitis did not significantly improve lameness or alter synovial fluid parameters. APS did lead to significant improvement in gross joint appearance and synovial membrane histology suggesting that APS may have disease-modifying effects.
Collapse
Affiliation(s)
- Alexandra Usimaki
- Department of Clinical Studies‐New Bolton CenterUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Sarah A. Ciamillo
- Department of Clinical Studies‐New Bolton CenterUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Dhvani Barot
- Department of Clinical Studies‐New Bolton CenterUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Renata L. Linardi
- Department of Clinical Studies‐New Bolton CenterUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Julie B. Engiles
- Department of Clinical Studies‐New Bolton CenterUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
- Department of PathobiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Kyla F. Ortved
- Department of Clinical Studies‐New Bolton CenterUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| |
Collapse
|
15
|
Ding L, Ren JY, Huang YF, Zhang JZ, Bai ZR, Leng Y, Tian JW, Wei J, Jin ML, Wang G, Li X, Qi X. Resistin upregulates fatty acid oxidation in synoviocytes of metabolic syndrome-associated knee osteoarthritis via CAP1/PKA/CREB to promote inflammation and catabolism. Arthritis Res Ther 2025; 27:99. [PMID: 40301946 PMCID: PMC12039190 DOI: 10.1186/s13075-025-03527-6] [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: 08/09/2024] [Accepted: 03/08/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Metabolic Syndrome (MetS), as a syndrome characterized by low-grade inflammation and energy metabolism disorders, is considered to be an important systemic risk factor for knee osteoarthritis (KOA). Our previous study showed that the protein level of serum resistin was positively correlated with the degree of metabolic disorder in MetS-OA. However, whether Resistin promotes the progression of KOA synovitis and the underlying mechanisms remain unclear. This study mainly investigateswhether there were metabolism disorder which promote inflammatory and catabolic phenotype in fibroblast-like synoviocytes (FLS) from KOA patients with MetS (MetS-KOA-FLS), and the roles and mechanisim of resistin in MetS-KOA-FLS. METHODS Comparative analysis of synovium and FLS from MetS-associated KOA (MetS-KOA) and non-MetS-associated KOA (nMetS-KOA) of females to detect the differences in inflammation, catabolism and glycolipid metabolism. Serum from MetS-KOA stimulated nMetS-KOA-FLS to detect the effect of MetS microenvironment on inflammation, catabolism and glycolipid metabolism of nMetS-KOA-FLS. Resistin stimulated MetS-KOA-FLS to explore the effect of resistin on inflammation and catabolism of MetS-KOA-FLS and its specific mechanism. RESULTS Compared with nMetS-KOA-FLS, MetS-KOA-FLS expressed higher inflammatory related factors, catabolic enzymes, and showed stronger adhesive and invasive ability. Resistin was found to be an important factor in the serum and internal environment of MetS-KOA patients, and it mediated the differences in fatty acid oxidation (FAO) between the two groups. Resistin activated the PKA/CREB pathway through CAP1 and upregulated FAO, promoting the inflammatory and catabolic phenotype of MetS-KOA-FLS. CONCLUSIONS This study clarifies the mechanism by which MetS causes synovitis from a metabolic perspective and provides new ideas for further research and treatment of MetS-KOA.
Collapse
Affiliation(s)
- Lu Ding
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Jin-Yi Ren
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Yi-Fan Huang
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Jian-Zeng Zhang
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China
| | - Zi-Ran Bai
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Yi Leng
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China
| | - Jun-Wei Tian
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China
| | - Jing Wei
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Min-Li Jin
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Guan Wang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, Liaoning, China.
| | - Xia Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, Liaoning, China.
| | - Xin Qi
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
16
|
Peng P, Zheng W, Liu Y, Huang J, Zhang B, Shen J, Cao J. Imrecoxib attenuates osteoarthritis by modulating synovial macrophage polarization through inactivating COX-2/PGE2 signaling pathway. Front Bioeng Biotechnol 2025; 13:1526092. [PMID: 40357333 PMCID: PMC12066670 DOI: 10.3389/fbioe.2025.1526092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Although biomaterials strategies have been regarded as a promising approach for the treatment of osteoarthritis (OA), identifying novel drugs to be delivered for modulate macrophage polarization is still unclear. As a commonly used non-steroidal anti-inflammatory drug for OA, Imrecoxib may be a novel drug to direct and sustain macrophage phenotype. However, the specific protective mechanism of Imrecoxib in OA remains unclear. This study aims to investigate whether Imrecoxib would treat OA by regulating synovial macrophage polarization. Methods The research involves constructing mouse destabilization of medial meniscus (DMM) model to assess the changes in pain, bone destruction, cartilage degeneration, and synovial macrophage phenotypes following Imrecoxib treatment. Additionally, the effects of macrophage conditioned medium (CM) pretreated with Imrecoxib on the chondrocyte apoptosis, inflammation and degeneration-related factor expression were evaluated. The role of COX-2/PGE2 signaling pathway in the macrophage phenotype changes was further investigated. Results We found that Imrecoxib alleviated pain, cartilage degeneration and synovitis, promoted polarization of M1 macrophages toward M2 phenotype in vivo and in vitro. In vitro experiments, Imrecoxib-CM protected chondrocyte by modulating macrophage polarization. Furthermore, Imrecoxib regulates macrophage polarization through the COX-2/PGE2 pathway. Conclusion This study unravels that Imrecoxib protects joint cartilage and attenuates osteoarthritis by modulating synovial macrophage polarization through inactivating COX-2/PGE2 signaling pathway, providing new drug delivery strategy for the clinical treatment of OA.
Collapse
Affiliation(s)
- Peng Peng
- Department of Sports injury and Arthroscopy, Tianjin University Tianjin Hospital, Tianjin, China
- Department of Neurosurgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanling Zheng
- Department of Dermatology and cosmetology, Minhang Hospital, Fudan University, Shanghai, China
| | - Yuchen Liu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingyuan Huang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bin Zhang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiawei Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jiangang Cao
- Department of Sports injury and Arthroscopy, Tianjin University Tianjin Hospital, Tianjin, China
| |
Collapse
|
17
|
Marcellin-Little DJ, Hulse DA, Huntingford JL, Grubb T, Brunke MW, Markley AP, Frank B. A proposed framework for practical multimodal management of osteoarthritis in growing dogs. Front Vet Sci 2025; 12:1565922. [PMID: 40357193 PMCID: PMC12067799 DOI: 10.3389/fvets.2025.1565922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoarthritis (OA) is a ubiquitous problem affecting dog joints, particularly the hip, elbow, stifle, and spine. OA most often results from developmental orthopedic problems such as hip dysplasia, elbow dysplasia, and patellar luxation and from injuries to the cranial cruciate ligament. Several management approaches have been proposed to manage OA, including steps to modulate growth, physical activity, and exercise, nutrition and nutritional supplementation, medications, physical rehabilitation, and surgical procedures. This article is the first in a series of articles that propose steps for practical OA management in dogs at various life stages. The review presented here focuses on growing dogs. The text describes the early pathophysiology and diagnosis of OA. The physical, nutritional, analgesic, and surgical management options of OA in growing dogs are presented. The application of these management options is described for three dogs. The overall approach to the management of OA in growing dogs is discussed.
Collapse
Affiliation(s)
- Denis J. Marcellin-Little
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Donald A. Hulse
- College of Veterinary Medicine, Texas A&M University, College Station, TX, United States
| | | | - Tamara Grubb
- Department of Veterinary Clinical Sciences, Washington State University, Pullman, WA, United States
| | | | | | - Bethany Frank
- Vetoquinol United States, Fort Worth, TX, United States
| |
Collapse
|
18
|
Hsieh WC, Hsu TS, Wu KW, Lai MZ. Therapeutic application of regulatory T cell in osteoarthritis. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025:S1684-1182(25)00083-0. [PMID: 40300967 DOI: 10.1016/j.jmii.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/07/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025]
Abstract
Regulatory T cells (Tregs) are the specific T cell population that suppress inflammatory immunity. Independent of their inhibitory activities, Tregs exhibit unique capacity to repair tissue damage. Rapid progresses are made in the processing and engineering of Tregs for clinical applications. Tregs have been used in the treatment of autoimmune diseases, transplantation rejection and graft-versus-host disease. Osteoarthritis is one of the major diseases that affect at least 600 million people worldwide. Osteoarthritis is characterized by physical erosion of cartilage, accompanied with chronic and low-grade inflammation. Tregs possess abilities to increase osteoclast differentiation and bone resorption, repair bone physical damage, and increase bone mass. Tregs are therefore candidate therapeutics for osteoarthritis for both inflammation resolution and tissue repairing. In this review, we will summarize the recent development in using Tregs in immunotherapy, and the potential of using Tregs in osteoarthritis.
Collapse
Affiliation(s)
- Wan-Chen Hsieh
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| | - Kuan-Wen Wu
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan.
| | - Ming-Zong Lai
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
19
|
Lin N, Song Z, Ma B, Wang P. LncRNA SNHG7 inhibits apoptosis and proliferation of osteoarthritis cells induced by IL-β through sponging miR-146b. Connect Tissue Res 2025:1-14. [PMID: 40207563 DOI: 10.1080/03008207.2025.2487470] [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: 05/20/2024] [Revised: 01/24/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE We probed the roles of SNHG7, miR-146b, PCBP1, and IL-β in the development of osteoarthritis (OA). MATERIALS AND METHODS OA models were established using anterior cruciate ligaments, and chondrocytes were obtained from mouse cartilage tissue. Cells were treated with 10 ng/ml Il-1β. RT-qPCR was used to detect the expression of SNHG7, miR-146b, PCBP1, and IL-β in tissues and cells. Safranin-O/Fast Green staining was performed to analyze the cartilage damage in each group of mice. RESULTS SNHG7 and PCBP1 expressions were down-regulated, and miR-146b expression was up-regulated in OA tissue and IL-1β-treated chondrocytes compared to normal cartilage tissue and chondrocytes. Forced SNHG7 expression improved cartilage structure, enhanced proliferative viability of chondrocytes, and inhibited apoptosis and IL-1β release in IL-1β-treated chondrocytes in OA mice. In contrast, miR-146b upregulation decreased proliferative viability and promoted apoptosis and IL-1β release in chondrocytes. Rescue assays showed that miR-146b attenuated the protective effects of SNHG7 on apoptosis and inflammation in IL-1β-treated chondrocytes, and activation of PCBP1 expression significantly inhibited the cytotoxic effects of miR-146b. Mechanistically, SNHG7 acted as a competitive endogenous RNA by targeting miR-146b to promote the expression of PCBP1. CONCLUSIONS This study confirms that SNHG7 inhibits IL-1β-mediated inflammatory responses in chondrocytes via the miR-146b/PCBP1 axis, thereby suppressing IL-1β-induced OA.
Collapse
Affiliation(s)
- Naikai Lin
- Department of Orthopedics, Hangzhou Yuhang District First People's Hospital, Zhejiang, China
| | - Zehui Song
- Department of Orthopedics, Hangzhou Yuhang District First People's Hospital, Zhejiang, China
| | - Bitao Ma
- Department of Traditional Chinese Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peng Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Chen R, Zheng S, Zhao X, Huang H, Xu Y, Qiu C, Li S, Liang X, Mao P, Yan Y, Lin Y, Song S, Cai W, Guan H, Yao Y, Zhu W, Shi X, Ganapathy V, Kou L. Metabolic reprogramming of macrophages by a nano-sized opsonization strategy to restore M1/M2 balance for osteoarthritis therapy. J Control Release 2025; 380:469-489. [PMID: 39921035 DOI: 10.1016/j.jconrel.2025.02.005] [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: 10/22/2024] [Revised: 01/26/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Osteoarthritis is a chronic and progressive joint disease accompanied by cartilage degeneration and synovial inflammation. It is associated with an imbalance of synovial macrophage M1/M2 ratio tilting more towards the pro-inflammatory M1 than the anti-inflammatory M2. The M1-macrophages rely on aerobic glycolysis for energy whereas the M2-macrophages derive energy from oxidative phosphorylation. Therefore, inhibiting aerobic glycolysis to induce metabolic reprogramming of macrophages and consequently promote the shift from M1 type to M2 type is a therapeutic strategy for osteoarthritis. Here we developed a macrophage-targeting strategy based on opsonization, using nanoparticles self-assembled to incorporate Chrysin (an anti-inflammatory flavonoid) and V-9302 (an inhibitor of glutamine uptake), and the outer layer modified by immunoglobulin IgG by electrostatic adsorption into IgG/Fe-CV NPs. In vitro studies showed that IgG/Fe-CV NPs effectively target M1 macrophages and inhibit HIF-1α and GLUT-1 essential for aerobic glycolysis and promote polarization from M1 to M2-type macrophages. In vivo, IgG/Fe-CV NPs inhibit inflammation and protect against cartilage damage. The metabolic reprogramming strategy with IgG/Fe-CV NPs to shift macrophage polarization from inflammatory to anti-inflammatory phenotype by inhibiting aerobic glycolysis and glutamine delivery may open up new avenues to treat osteoarthritis.
Collapse
Affiliation(s)
- Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Shimin Zheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xinyu Zhao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Huirong Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yitianhe Xu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenyu Qiu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shengjie Li
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xindan Liang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Pengfei Mao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yuqi Yan
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yinhao Lin
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shengnan Song
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wenjing Cai
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Haoxiong Guan
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yinsha Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wanling Zhu
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China.
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
21
|
Gao T, Chen T, Ai C, Gu Y, Wang Y, Zhou X, Zhao C. The Causal Relationship Between Zinc and Osteoarthritis: A Two-Sample Mendelian Randomization Study. Biol Trace Elem Res 2025:10.1007/s12011-025-04611-3. [PMID: 40195255 DOI: 10.1007/s12011-025-04611-3] [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: 02/02/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
There is a clear relationship between osteoarthritis (OA) and micronutrients. Excessive accumulation of micronutrients may play a negative role in aggravating the symptoms of OA. This study aims to sort out the causal relationship between micronutrients (zinc, copper, magnesium, vitamins A, C, E, D, B6, and B12, folic acid, iron, carotene, selenium, calcium, and potassium) and OA. This study used Mendelian randomization (MR) to combining the causal relationship between micronutrients and the risk of OA. Micronutrient-related variants were extracted from a large-scale genome-wide association study (GWAS) database of circulating micronutrients in European populations. Outcome data were from the FINNGEN meta-analysis of OA in participants of European ancestry from the FinnGen Biobank in Finland. The primary analysis was performed using the inverse-variance weighted (IVW) method, and a series of sensitivity analyses and multi-dimensionality analyses were conducted to detect possible violations of the MR assumptions. This study used the IVW method to analyze the causal relationship between 15 micronutrients and OA. The results showed that copper (P = 0.535), selenium (P = 0.463), folic acid (P = 0.664), carotene (P = 0.706), potassium (P = 0.839), vitamin D (P = 0.941), vitamin C (P = 0.928), vitamin B12 (P = 0.859), iron (P = 0.496), vitamin E (P = 0.678), magnesium (P = 0.934), vitamin B6 (P = 0.027), calcium (P = 0.743), and vitamin A (P = 0.368) had no significant causal relationship with OA. Among them, vitamin B6 showed P < 0.05 in the pleiotropy test, indicating the presence of pleiotropy. In contrast, zinc exhibited a significant causal relationship with OA (P < 0.001, OR 95% CI = 1.044 [1.021-1.067]), with sensitivity analyses further validating the robustness and reliability of this finding. This study reveals a causal relationship between zinc and OA, identifying zinc as a risk factor for OA. It provides evidence of causality between zinc and OA, offering novel insights for clinical research, diagnosis, and treatment of OA.
Collapse
Affiliation(s)
- TianQi Gao
- Changchun University of Chinese Medicine, Changchun, China
| | - TianYang Chen
- Changchun University of Chinese Medicine, Changchun, China
| | - ChengLong Ai
- Changchun University of Chinese Medicine, Changchun, China
| | - Yan Gu
- Changchun University of Chinese Medicine, Changchun, China
| | - YunPeng Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - XiaoLing Zhou
- Department of Orthopedics, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China.
| | - ChangWei Zhao
- Department of Orthopedics, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China.
| |
Collapse
|
22
|
Castanheira CIGD, Anderson JR, Clarke EJ, Hackl M, James V, Clegg PD, Peffers MJ. Extracellular Vesicle-Derived microRNA Crosstalk Between Equine Chondrocytes and Synoviocytes-An In Vitro Approach. Int J Mol Sci 2025; 26:3353. [PMID: 40244190 PMCID: PMC11989968 DOI: 10.3390/ijms26073353] [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: 02/17/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
This study describes a novel technique to analyze the extracellular vesicle (EV)-derived microRNA (miRNA) crosstalk between equine chondrocytes and synoviocytes. Donor cells (chondrocytes, n = 8; synoviocytes, n = 9) were labelled with 5-ethynyl uridine (5-EU); EVs were isolated from culture media and incubated with recipient cells (chondrocytes [n = 5] were incubated with synoviocyte-derived EVs, and synoviocytes [n = 4] were incubated with chondrocyte-derived EVs). Total RNA was extracted from recipient cells; the 5-EU-labelled RNA was recovered and sequenced. Differential expression analysis, pathway analysis, and miRNA target prediction were performed. Overall, 198 and 213 miRNAs were identified in recipient synoviocytes and chondrocytes, respectively. The top five most abundant miRNAs were similar for synoviocytes and chondrocytes (eca-miR-21, eca-miR-221, eca-miR-222, eca-miR-100, eca-miR-26a), and appeared to be linked to joint homeostasis. There were nine differentially expressed (p < 0.05) miRNAs (eca-miR-27b, eca-miR-23b, eca-miR-31, eca-miR-191a, eca-miR-199a-5p, eca-miR-143, eca-miR-21, eca-miR-181a, and eca-miR-181b) between chondrocytes and synoviocytes, which appeared to be linked to migration of cells, apoptosis, cell viability of connective tissue cell, and inflammation. In conclusion, the reported technique was effective in recovering and characterizing the EV-derived miRNA crosstalk between equine chondrocytes and synoviocytes and allowed for the identification of EV-communicated miRNA patterns potentially related to cell viability, inflammation, and joint homeostasis.
Collapse
Affiliation(s)
- Catarina I. G. D. Castanheira
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK (J.R.A.); (E.J.C.); (P.D.C.)
| | - James R. Anderson
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK (J.R.A.); (E.J.C.); (P.D.C.)
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RP, UK
| | - Emily J. Clarke
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK (J.R.A.); (E.J.C.); (P.D.C.)
| | | | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham LE12 5RD, UK;
| | - Peter D. Clegg
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK (J.R.A.); (E.J.C.); (P.D.C.)
| | - Mandy J. Peffers
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK (J.R.A.); (E.J.C.); (P.D.C.)
| |
Collapse
|
23
|
Gao C, Feng Y, Liu S, Chen B, Ding M, Du D, Zhang W, Wilson DA, Tu Y, Peng F. Light-Driven Artificial Cell Micromotors for Degenerative Knee Osteoarthritis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416349. [PMID: 40025988 DOI: 10.1002/adma.202416349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/02/2025] [Indexed: 03/04/2025]
Abstract
Combining artificial cellular compartmentalization and intelligent motion benefits of micro/nanomotors, light is used as energy input to construct an artificial cell-based micromotor capable of photosynthetic anabolism and intelligent directional movement. This system is assembled from phospholipids functionalized with F-ATP synthase and molybdenum disulfide (MoS2) nanoparticles (Vesical@MoS2-ATPase). The underlying mechanism involves the generation of protons (H+) through photo-hydrolysis of MoS2 nanoparticles within vesicles, which generates a local electroosmotic flow inside the vesicles and drives the negatively charged MoS2 toward light. The established proton gradient across the phospholipid membrane, in turn, drives the ATP synthase to catalyze ATP production. Both in vitro and in vivo models demonstrate that the micromotor can elevate local intracellular ATP levels upon light and improve the metabolism of denatured chondrocytes. This cell mimicry, with capabilities of migration and biosynthesis, emerges as a promising platform for the next generation of functional bio-interface.
Collapse
Affiliation(s)
- Chao Gao
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Ye Feng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Suyi Liu
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Bin Chen
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Miaomiao Ding
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Dailing Du
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Wenjing Zhang
- School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Daniela A Wilson
- Institute for Molecules and Materials, Radboud University, Nijmegen, AJ 6525, The Netherlands
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| |
Collapse
|
24
|
Chen Y, Xu N, Zhang W, Wang Y, Su T, Zhou Y, Xu J. FSH enhances the inflammatory response of macrophages in the knee joint possibly through the NFκB pathway. FEBS Open Bio 2025; 15:622-633. [PMID: 39801258 PMCID: PMC11961395 DOI: 10.1002/2211-5463.13959] [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: 03/24/2024] [Revised: 10/22/2024] [Accepted: 12/13/2024] [Indexed: 04/03/2025] Open
Abstract
Previous studies have suggested that women with higher follicle-stimulating hormone (FSH) levels have a greater incidence of osteoarthritis (OA) compared to women with lower FSH despite normal estrogen levels. Our previous studies also showed that FSH has a negative effect on cartilage in postmenopausal OA. However, no studies have investigated the effect of FSH on the synovium. Here, we showed that the FSH receptor (FSHR) is expressed on RAW264.7 cells and BMDM (Bone Marrow-Derived Macrophages), and found that FSH stimulation promotes the production and secretion of inflammatory cytokines in synovial macrophages. In RAW264.7 cells, FSH stimulation enhances phosphorylation and nuclear translocation of P65, suggesting the activation of NFκB signaling, while the knockdown of FSHR eliminates the proinflammatory effect of FSH. To further validate these results, we used an ovariectomy mouse model supplemented with FSH and estrogen, and a mouse model with FSH neutralization. We noted that FSHR was expressed on mouse synovial joint membranes. Furthermore, in ovariectomy mice supplemented with estrogen and treated with FSH, synovial macrophages were significantly increased, while the opposite was the case in the FSH neutralizing group, which suggest that FSH triggers an inflammatory response in the synovial tissue in mice. Taken together, our results indicate that FSH is an important regulator in synovial inflammation via NFκB signaling activation and, to some extent, appears to accelerate the development of osteoarthritis.
Collapse
Affiliation(s)
- Yu Chen
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Na Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Wen‐wen Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Yan Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Tong Su
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Yan‐man Zhou
- Department of NephrologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Jin Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| |
Collapse
|
25
|
Gao K, Huang Z, Liao Z, Wang Y, Chen D. Machine learning analysis of FOSL2 and RHoBTB1 as central immunological regulators in knee osteoarthritis synovium. J Int Med Res 2025; 53:3000605251333646. [PMID: 40287984 PMCID: PMC12035077 DOI: 10.1177/03000605251333646] [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: 07/25/2024] [Accepted: 02/25/2025] [Indexed: 04/29/2025] Open
Abstract
BackgroundKnee osteoarthritis is a debilitating disease with a complex pathogenesis. Synovitis, which refers to inflammation of the synovial membrane surrounding the joint, is believed to play an important role in the development and progression of knee osteoarthritis. To better understand the molecular mechanisms underlying knee osteoarthritis, we conducted a comprehensive analysis of gene expression in knee osteoarthritis synovium using machine learning.MethodsDifferentially expressed genes between knee osteoarthritis and control synovial tissues were analyzed using the GSE55235 dataset. We employed several machine learning algorithms, including least absolute shrinkage and selection operator and support vector machine-recursive feature elimination, to screen for key genes. Then, we validated the key genes using an external dataset (GSE51588) and an in vitro knee osteoarthritis animal model. CIBERSORT was used to compare immune cell infiltration levels between knee osteoarthritis and control synovial tissues and determine their relationship with the key genes. Finally, we performed a Connectivity Map analysis to screen for potential small-molecule compounds. Moreover, we conducted single-cell RNA sequencing analysis using knee joint tissues to annotate different subtypes of cells.ResultsA total of 930 differentially expressed genes were identified. Least absolute shrinkage and selection operator regression and support vector machine-recursive feature elimination identified FOSL2 and RHoBTB1 as key genes. The expression levels of both genes were further validated in the GSE51588 dataset as well as verified through an in vitro experiment involving a knee osteoarthritis mouse model. Multiple significant correlation pairs were found between the immune cell infiltration levels. We unveiled the genetic basis of knee osteoarthritis using genome-wide association study and specific signaling pathways through gene set enrichment analysis. The GeneCards database was used to obtain 3032 pathogenic genes associated with knee osteoarthritis, and we found that RHoBTB1 expression was significantly negatively correlated and FOSL2 expression was significantly positively correlated with interleukin-1β expression. We predicted several small-molecule compounds based on Connectivity Map analysis. Finally, single-cell RNA sequencing analysis revealed the expression levels of the two key genes in chondrocytes and tissue stem cells.ConclusionFOSL2 and RHoBTB1 may play key roles in the pathogenesis of knee osteoarthritis, exhibiting correlations with immune cell infiltration levels. These findings indicate that these genes have potential as therapeutic targets. However, further research and validation are necessary to confirm their exact roles and therapeutic potential in knee osteoarthritis.
Collapse
Affiliation(s)
- Kun Gao
- Department of Orthopedics, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhenyu Huang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhouwei Liao
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yanfei Wang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Dayu Chen
- Department of Orthopedics, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
26
|
Zhang H, Wei X, Liu W, Leng H, Shen Q, Wan X, Xu X, Zheng X. Latent class analysis identifies distinct pain phenotypes in newly diagnosed systemic juvenile idiopathic arthritis. Arthritis Res Ther 2025; 27:71. [PMID: 40165337 PMCID: PMC11956179 DOI: 10.1186/s13075-025-03534-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Patients with systemic juvenile idiopathic arthritis (sJIA) exhibit highly heterogeneous pain manifestations, which significantly impact their quality of life and disease prognosis. An understanding of the pain phenotypes for this disorder and their influencing factors is crucial for individualized pain management. OBJECTIVE To explore the pain phenotypes of newly diagnosed sJIA patients via latent class analysis (LCA), analyse the influencing factors of these phenotypes, and evaluate the impacts of different pain phenotypes on short-term inpatient outcomes. METHODS A retrospective cohort study was conducted by collecting the electronic health records of 165 patients who were first diagnosed with sJIA at the Children's Hospital of Chongqing Medical University from January 2018 to July 2024. Patient pain characteristics, laboratory indicators, and inpatient outcome data were extracted. LCA was used to identify pain phenotypes, and multivariate logistic regression was used to analyse the influencing factors. The Lanza-Tan-Bray method and the data combination analysis technique were applied to evaluate the relationships between pain phenotypes and clinical outcomes. RESULTS LCA categorized the pain phenotypes of sJIA patients into three distinct classes, including (1) Class 1: inflammation-related moderate to severe pain with functional impairment (53.9% of patients); (2) Class 2: mild intermittent pain with extra-articular symptoms (19.4% of patients); and (3) Class 3: no joint pain with mild functional impairment (26.7% of patients). The analysis revealed that age (P = 0.023) and serum IL-10 levels (P = 0.047) were significant factors influencing pain phenotypes. Significant differences were observed among different pain phenotypes in terms of hospital stay duration, intrahospital department transfer rates, and pain status at discharge. CONCLUSION Pain in sJIA patients can be classified into three distinct phenotypes, which are influenced by factors such as age and IL-10 levels. The identification of these pain phenotypes has important clinical significance for developing individualized pain management strategies.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Nursing, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqiong Wei
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Liu
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hongyao Leng
- Department of Nursing, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qiao Shen
- Department of Nursing, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Wan
- Department of Nursing, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ximing Xu
- Big Data Center for Children's Medical Care, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Xianlan Zheng
- Department of Nursing, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
27
|
Zhu Z, Tu B, Peng C, Xu X, Lu P, Ning R. Integrated bioinformatics and clinical data identify three novel biomarkers for osteoarthritis diagnosis and synovial immune. Sci Rep 2025; 15:10987. [PMID: 40164659 PMCID: PMC11958655 DOI: 10.1038/s41598-025-95837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease that can be aggravated by synovitis and synovial immune disorders (SID). However, the role of synovial SID-related genes in OA synovium remains poorly understood. OA synovial and peripheral blood datasets were obtained from the GEO database ( https://www.ncbi.nlm.nih.gov/ ). Immune-related genes ( https://reactome.org/ ) showing differential expression in peripheral blood were identified as immune disorder genes. Subsequently, differentially expressed immune disorder genes in OA synovium were further identified as SID genes. The Venn diagram, random forest, SVM-RFE algorithm, and multivariate analysis were employed to determine SID-related hub genes in OA synovium. Using the identified hub genes, we constructed and validated a diagnostic model for predicting OA occurrence. The correlation between hub gene expression and immune-related modules was explored using CIBERSORT and MCP-counter analyses. We identified three SID-related hub genes (ACAT1, SPHK1, and ACACB) in OA synovium. The diagnostic model incorporating these hub genes demonstrated reliable predictive accuracy (AUC = 0.939). Through qPCR analysis, we quantitated the expression levels of the hub genes and confirmed that three hub genes could serve as novel biomarkers for OA patients (AUC = 0.960). Furthermore, we observed a significant correlation between the expression of these hub genes and immune cell infiltration, as well as inflammatory cytokine levels in OA synovium. Our findings suggest that three SID-related hub genes have the potential to serve as diagnostic biomarkers for OA patients. These genes are associated with immune disorder and contribute to immune alterations within the OA synovium.
Collapse
Affiliation(s)
- Zheng Zhu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Bizhi Tu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Cheng Peng
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Xun Xu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Peizhi Lu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Rende Ning
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China.
| |
Collapse
|
28
|
Lai P, Ma Y, Sang W, Zhou Q, Chen H, Wang C, Yin J, Wang T, Zhu L, Zhou X, He C, Ma J. Reprogramming Macrophage Phenotype Using a Reactive Oxygen Species-Responsive Liposome Delivery System for Inflammation Microenvironment Remodeling and Osteoarthritis Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:17932-17947. [PMID: 40094857 DOI: 10.1021/acsami.4c19160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The progression of osteoarthritis (OA) is closely linked to synovial inflammation caused by an imbalance between M1 and M2 macrophages. To tackle this problem, we developed a liposome responsive to reactive oxygen species (ROS), modified with folic acid ligands to target M1-polarized macrophages, and loaded with the anti-inflammatory agent dimethyl fumarate (DMF). This liposome-based drug delivery system was designed to reprogram macrophage phenotype to remodel the inflammatory microenvironment in the joint cavity and alleviate OA degeneration. The liposome we prepared had a suitable size and negative zeta potential, with uniform size, good stability in aqueous solution, and excellent biocompatibility. Laboratory tests showed that these DMF-filled liposomes notably decreased high levels of ROS in M1-type macrophages and shifted macrophage polarization via the Nrf2/HO-1 pathway, which in turn lessened inflammation in chondrocytes and averted their apoptosis. Additionally, animal studies demonstrated that liposomes containing DMF exhibited notable anti-inflammatory properties, significantly reduced synovial inflammation, safeguarded injured cartilage, reversed changes in subchondral bone, and effectively slowed the progression of osteoarthritis in a mouse model induced by anterior cruciate ligament transection (ACLT). Therefore, ROS-responsive liposomes targeting M1-polarized macrophages represent a promising and valuable approach for OA treatment.
Collapse
Affiliation(s)
- Peng Lai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yichao Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Weilin Sang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Qiang Zhou
- Department of Otolaryngology, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital), Wenzhou 325200, China
| | - Hongjie Chen
- The School of Medicine, Nankai University, Tianjin 300071, China
| | - Cong Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jian Yin
- Department of Orthopedics, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing 211100, China
| | - Tao Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Libo Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiaojun Zhou
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Chuanglong He
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
29
|
Yao Z, Li Y, Mai H, Wang Z, Zhang H, Cai D, Wang X. Comprehensive multiomics analysis identifies PYCARD as a key pyroptosis-related gene in osteoarthritis synovial macrophages. Front Immunol 2025; 16:1558139. [PMID: 40196125 PMCID: PMC11973068 DOI: 10.3389/fimmu.2025.1558139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Background Osteoarthritis (OA) is a chronic joint disease that significantly impairs quality of life. Synovitis plays a pivotal role in OA progression, and pyroptosis, a form of programmed cell death associated with innate immune inflammation, may contribute to the pathogenesis of OA synovitis. Nevertheless, the precise role of pyroptosis in OA pathogenesis remains poorly understood. Methods We performed an analysis of bulk RNA sequencing data to examine the expression profiles of pyroptosis-related genes in the OA synovium. A LASSO-Cox regression model was employed to identify pivotal genes. Single-cell RNA sequencing data were used to validate the expression of these genes in specific synovial cell clusters. Differentially expressed genes (DEGs) in macrophages with high or low expression levels of core genes were subjected to enrichment analysis. A protein-protein interaction (PPI) network was constructed to identify hub genes, and potential therapeutic compounds were predicted. Consensus clustering analysis was performed to examine the correlations between hub genes and disease status. After identifying PYCARD as the core pyroptosis gene in OA macrophages, we assessed the expression levels of PYCARD in the OA synovium and validated the expression of PYCARD and its related core genes in M1 macrophages. Results A total of twenty pyroptosis-related DEGs were identified, and six core genes were selected through LASSO regression. PYCARD was identified as the key pyroptosis gene in macrophages. Furthermore, 57 therapeutic compounds targeting these genes were predicted. Validation confirmed the upregulation of PYCARD in the OA synovium and M1 macrophages. Conclusion PYCARD was identified as the core pyroptosis gene in OA macrophages, and 57 potential therapeutic compounds were identified. This study offers valuable insights into potential treatment targets for OA.
Collapse
Affiliation(s)
- Zihao Yao
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Yuexin Li
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Hanwen Mai
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Zhuolun Wang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Haiyan Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Daozhang Cai
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Xiangjiang Wang
- Department of Orthopedics, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
30
|
Griffith JL, Joseph J, Jensen A, Banks S, Allen KD. Using deep-learning based segmentation to enable spatial evaluation of knee osteoarthritis (SEKO) in rodent models. Osteoarthritis Cartilage 2025:S1063-4584(25)00867-2. [PMID: 40139644 DOI: 10.1016/j.joca.2025.02.787] [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: 07/25/2024] [Revised: 01/21/2025] [Accepted: 02/20/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE In preclinical models of osteoarthritis (OA), histology is commonly used to evaluate joint remodeling. The current study introduces a deep learning driven histological analysis pipeline for the spatial evaluation of knee osteoarthritis (SEKO) focused on quantifying and visualizing joint remodeling in the medial compartment of rodent knees. METHODS The SEKO pipeline contains both segmentation and visualization tools. For segmentation, two separate convolutional neural network architectures, HRNet and U-Net, were considered for identifying multiple regions of interest. Following segmentation, SEKO calculates multiple morphometric and location dependent measures to summarize joint-level changes. Additionally, SEKO generates probabilistic heat maps for visualization of the spatial aspects of joint remodeling. RESULTS SEKO incorporated the U-NET architecture - due to its higher prediction accuracy - and identified similar cartilage loss changes that were reported using by-hand segmentation in prior work. Additionally, SEKO enabled the detection of changes in subchondral bone area and location dependent bone remodeling. SEKO also enabled visualization of spatial changes in cartilage thinning and bone remodeling using probabilistic heat maps. CONCLUSION The SEKO pipeline offers the potential for objective comparison of OA progression and therapeutic interventions through visualization of spatial and morphometric changes. SEKO is provided as an open-source tool for the OA research community, facilitating collaborative research efforts and comprehensive analysis of knee joint histology.
Collapse
Affiliation(s)
- Jacob L Griffith
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Pain Research & Intervention Center of Excellence (PRICE), University of Florida, Gainesville, FL, USA
| | - Justin Joseph
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Andrew Jensen
- Department of Mechanical and Aerospace Engineering at the University of Florida, Gainesville, FL, USA
| | - Scott Banks
- Department of Mechanical and Aerospace Engineering at the University of Florida, Gainesville, FL, USA; Department of Orthopaedic Surgery and Sports Medicine, University of Florida, Gainesville, FL, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Pain Research & Intervention Center of Excellence (PRICE), University of Florida, Gainesville, FL, USA; Department of Mechanical and Aerospace Engineering at the University of Florida, Gainesville, FL, USA; Department of Orthopaedic Surgery and Sports Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
31
|
Liao Q, Chen J, Liu G. Low intensity pulsed ultrasound alleviates synovial fibrosis in osteoarthritis via the PI3K/AKT pathway. Sci Rep 2025; 15:9644. [PMID: 40113833 PMCID: PMC11926212 DOI: 10.1038/s41598-025-92413-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
Previous studies have reported that low-intensity pulsed ultrasound (LIPUS) can alleviate cartilage degradation in osteoarthritis (OA). However, the functions and mechanisms of LIPUS in synovial fibrosis with OA require further study. To investigate the role of the PI3K/AKT signaling pathway in synovial fibrosis and in LIPUS treatment in synovial fibrosis, a TGF-β stimulated rat FLS cell model and a rat OA animal model based on anterior cruciate ligament transection (ACLT) and partial medial meniscectomy (MMx) were used. The results revealed that LIPUS delayed the progression of OA. Masson staining revealed that LIPUS reduced the collagen deposition of synovial tissue in OA rats. Correspondingly, immunofluorescence demonstrated that LIPUS significantly downregulated the expression of α-SMA, Col1a1 and Col3a1 in OA rats. Moreover, TGF-β stimulation upregulated fibrosis markers at the mRNA and protein levels in FLS, as well as increased phosphorylation-dependent activation of the PI3K/Akt pathway. 740Y-P was found to promote the fibrotic change of FLS induced by TGF-β, but LY294002 reduced its expression. However, LIPUS inhibits the fibrotic change and activation of the PI3K/Akt pathway in FLS under stimulation of TGF-β. In conclusion, LIPUS alleviates synovial fibrosis by blocking the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Qing Liao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China
| | - Jun Chen
- Department of Rehabilitation Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, 510000, China
- Taihe Hospital, Hubei University of Medicine, Shiyan City, 442000, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
32
|
Tepecik T, Gedik E. Efficacy of adjunctive injectable platelet-rich fibrin as a first-line treatment in temporomandibular joint osteoarthritis: a retrospective cohort study. Int J Oral Maxillofac Surg 2025:S0901-5027(25)00078-5. [PMID: 40082134 DOI: 10.1016/j.ijom.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
This study was performed to compare the outcomes of adjunctive injections (injectable platelet-rich fibrin (iPRF), hyaluronic acid (HA)) and arthrocentesis only (AO) in the treatment of temporomandibular joint osteoarthritis. The study included 127 female patients (mean age 52.3 years). None of them had undergone prior occlusal splint therapy. The pain reduction (visual analogue scale) at 12 months post-treatment was the primary outcome variable, while maximum inter-incisal opening (MIO) at all follow-ups and pain at 1 and 6 months of follow-up were secondary outcome variables. All treatment groups showed significant improvements in pain and MIO at all postoperative follow-ups compared to baseline (P < 0.001). iPRF and HA resulted in significantly better pain relief than AO at all follow-ups (P < 0.01). There was no significant difference in pain alleviation between iPRF and HA. No differences were observed among the three groups in jaw mobility (MIO) at any follow-up. Since iPRF did not provide additional benefits over HA, the two treatments may be considered as alternatives, depending on cost considerations. Of note, the treatment objectives were achieved even without previous occlusal splint use.
Collapse
Affiliation(s)
- T Tepecik
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Health Sciences, Uskudar, Istanbul, Turkey.
| | - E Gedik
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Health Sciences, Uskudar, Istanbul, Turkey.
| |
Collapse
|
33
|
Arjmanddoust Z, Nazari A, Moezy A. Efficacy of two doses of intra-articular ozone therapy for pain and functional mobility in knee osteoarthritis: a double-blind randomized trial. Adv Rheumatol 2025; 65:11. [PMID: 40050950 DOI: 10.1186/s42358-025-00443-w] [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: 12/22/2024] [Accepted: 03/02/2025] [Indexed: 05/13/2025] Open
Abstract
AIM This double-blind, trial sought to assess the effectiveness of intra-articular ozone therapy at concentrations of 20 µg/mL and 40 µg/mL in managing pain and enhancing functional mobility in patients with knee osteoarthritis (KOA). METHOD This parallel, three-arm randomized controlled trial, conducted between 2022 and 2023, included 59 knee osteoarthritis (KOA) patients randomly allocated to one of three groups: the first group received 40 µg/mL ozone therapy, the second group received 20 µg/mL ozone therapy, and the control group received oxygen. Functional mobility was assessed through the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), knee flexion range of motion (FROM), the Timed Up and Go (TUG) test, and the six-minute walk test (6MWT). Pain intensity was measured using the Visual Analog Scale (VAS) and the pain subscale of the WOMAC. Intra-articular injections were administered weekly for four consecutive weeks, with assessments conducted pre-treatment, and at two weeks, one month, and two months post-intervention. One-way ANOVA was employed for normally distributed quantitative data, while the Kruskal-Wallis test was utilized for non-normally distributed data. Qualitative variables were analyzed using the Chi-squared or Fisher's exact test, as appropriate. RESULTS The groups receiving intra-articular ozone therapy exhibited notable reductions in mean VAS scores and improvement in functional mobility variables when compared to the control group (p < 0.05). However, post-hoc analysis indicated no statistically significant differences between the 40 µg/mL and 20 µg/mL ozone therapy groups regarding these parameters (VAS, FROM, TUG, 6MWT, or WOMAC scores) (p > 0.05). CONCLUSION Both 20 µg/mL and 40 µg/mL doses of intra-articular ozone therapy prove to be effective in reducing pain and enhancing functional mobility in patients with knee osteoarthritis (KOA). Nevertheless, there was no significant difference in the efficacy between the two ozone concentrations. TRIAL REGISTRATION The trial is registered on us ClinicalTrials.gov in 2024-05-01 with the following ID code NCT06088706.
Collapse
Affiliation(s)
- Zahra Arjmanddoust
- Department of Sports and Exercise Medicine, School of Medicine, Hazrate-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Nazari
- Department of Sports and Exercise Medicine, School of Medicine, Hazrate-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Azar Moezy
- Department of Sports and Exercise Medicine, School of Medicine, Hazrate-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Campbell TM, Feibel R, Dilworth J, Laneuville O, Trudel G. Capsular stem cell function and tissue composition are associated with symptoms and radiographic severity in people with knee osteoarthritis. Ther Adv Musculoskelet Dis 2025; 17:1759720X251321941. [PMID: 40041009 PMCID: PMC11877474 DOI: 10.1177/1759720x251321941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/04/2025] [Indexed: 03/06/2025] Open
Abstract
Background Osteoarthritis (OA) is associated with lost range of motion in the affected joint(s). Evidence suggests that this may be due to increased activity of posterior capsule fibroblasts, cells in turn derived from mesenchymal stromal cells (MSCs). Objectives To test the hypotheses that (1) MSCs are more numerous in the posterior capsule of patients with knee flexion contracture (FC) and (2) in OA participants with knee FC, the MSC population in the posterior capsule differentiates toward a fibrotic phenotype. In order to complete these objectives, we looked for associations between capsule histologic and MSC outcomes with clinical outcomes. Design Cross-sectional translational research design using data from the Ottawa Knee Osteoarthritis (OKOA) database. Methods A total of 71 OKOA database participants and their relevant clinical and laboratory outcomes were included. Associations were first tested with bivariate correlation, then for p < 0.10, tested using a linear model. Results No lab-based differences between FC and no-FC groups we discovered. In the posterior capsule, there was an association between knee flexion and adipogenic capacity (p = 0.001), osteogenic capacity (p < 0.001), KL grade and percent "other" (mainly neurovascular) tissue (p = 0.039), visual analog scale pain, and percent fibrous tissue (p = 0.014). For the anterior capsule, there was an association between knee flexion (p = 0.002) and extension (p = 0.005) with MSC enumeration, KL grade with MSC fibrogenic capacity (p = 0.002), and Knee Injury and Osteoarthritis Outcome Score quality of life with chondrogenic capacity (p < 0.001). Conclusion Joint capsule composition, MSC enumeration, and function were associated with important clinical OA outcomes. These findings suggest that the entire joint capsule may play an important role in OA-related morbidity and progression and could represent an underappreciated target for OA treatment.
Collapse
Affiliation(s)
- T. Mark Campbell
- Department of Physical Medicine and Rehabilitation, Elisabeth Bruyère Hospital, 43 Bruyère Street, Ottawa, Ontario K1N 5C8, Canada
| | - Robert Feibel
- Division of Orthopedic Surgery, Department of Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Jeffrey Dilworth
- Department of Cellular and Molecular Biology, University of Wisconsin–Madison, Madison, WI, USA
| | - Odette Laneuville
- Bone and Joint Research Laboratory, Department of Cellular and Molecular Medicine, Ottawa Hospital Research Institute, Canada
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Guy Trudel
- Bone and Joint Research Laboratory, Department of Cellular and Molecular Medicine, Ottawa Hospital Research Institute, Canada
- Division of Physical Medicine and Rehabilitation, Faculty of Medicine, Department of Medicine, University of Ottawa, Canada
- Division of Physical Medicine and Rehabilitation, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| |
Collapse
|
35
|
Fukui D, Nishiyama D, Yamanaka M, Tamai H, Nishio N, Kawakami M, Yamada H. Development of a Novel Rat Knee Osteoarthritis Model Induced by Medial Meniscus Extrusion. Cartilage 2025; 16:108-117. [PMID: 37837194 PMCID: PMC11744626 DOI: 10.1177/19476035231205680] [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: 12/12/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVE The medial meniscus extrusion (MME) is associated with increased stress on the knee joint, which leads to cartilage degeneration. To evaluate the etiology of knee osteoarthritis, it is extremely important to create animal models of the disease that more closely resemble actual clinical conditions in terms of symptomatology, molecular biology, and histology. This study aimed to create a clinically relevant model of MME in rats. DESIGN Behavioral, molecular biological, and histological changes in the newly developed rat MME model were compared with those in sham and medial meniscus transection and medial collateral ligament transection (MMT) models to examine the characteristics of this model. RESULTS In the MME rat model, behavioral evaluation shows abnormalities in gait compared with the other 2 groups, and molecular biological evaluation of the infrapatellar synovia of rats shows that gene expression of inflammatory cytokines, matrix-degrading enzymes, and pain-related nerve growth factor was increased compared with the sham group. Furthermore, histological evaluation reveals that cartilage degeneration was the most severe in the MME group. CONCLUSIONS The newly developed MME model reproduced the characteristic pathology of MME in clinical practice, such as severe pain, inflammation, and rapid progression of osteoarthritis. The MME model, which might more closely mimic human knee osteoarthritis (OA), could be a useful model for elucidating the pathophysiology and considering therapeutic management for knee OA.
Collapse
Affiliation(s)
- Daisuke Fukui
- Department of Orthopedic Surgery, Wakayama Medical University, Wakayama, Japan
| | - Daisuke Nishiyama
- Department of Orthopedic Surgery, Wakayama Medical University, Wakayama, Japan
| | - Manabu Yamanaka
- Department of Orthopedic Surgery, Wakayama Medical University, Wakayama, Japan
| | - Hidenobu Tamai
- Department of Orthopedic Surgery, Wakayama Medical University, Wakayama, Japan
| | - Naoko Nishio
- Department of Orthopedic Surgery, Wakayama Medical University, Wakayama, Japan
| | - Mamoru Kawakami
- Department of Orthopedic Surgery, Saiseikai Wakayama Hospital, Wakayama, Japan
| | - Hiroshi Yamada
- Department of Orthopedic Surgery, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
36
|
Wang X, Xu L, Wu Z, Lou L, Xia C, Miao H, Dai J, Fei W, Wang J. Exosomes of stem cells: a potential frontier in the treatment of osteoarthritis. PRECISION CLINICAL MEDICINE 2025; 8:pbae032. [PMID: 39781279 PMCID: PMC11705996 DOI: 10.1093/pcmedi/pbae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025] Open
Abstract
The aging population has led to a global issue of osteoarthritis (OA), which not only impacts the quality of life for patients but also poses a significant economic burden on society. While biotherapy offers hope for OA treatment, currently available treatments are unable to delay or prevent the onset or progression of OA. Recent studies have shown that as nanoscale bioactive substances that mediate cell communication, exosomes from stem cell sources have led to some breakthroughs in the treatment of OA and have important clinical significance. This paper summarizes the mechanism and function of stem cell exosomes in delaying OA and looks forward to the development prospects and challenges of exosomes.
Collapse
Affiliation(s)
- Xiaofei Wang
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Lei Xu
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Zhimin Wu
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Linbing Lou
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Cunyi Xia
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Wenyong Fei
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jingcheng Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
37
|
Zou Z, Hu W, Kang F, Xu Z, Li Y, Zhang J, Li J, Zhang Y, Dong S. Interplay between lipid dysregulation and ferroptosis in chondrocytes and the targeted therapy effect of metformin on osteoarthritis. J Adv Res 2025; 69:515-529. [PMID: 38621621 PMCID: PMC11954841 DOI: 10.1016/j.jare.2024.04.012] [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: 01/25/2024] [Revised: 04/03/2024] [Accepted: 04/13/2024] [Indexed: 04/17/2024] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is a devastating whole-joint disease affecting a large population worldwide; the role of lipid dysregulation in OA and mechanisms underlying targeted therapy effect of lipid-lowering metformin on OA remains poorly defined. OBJECTIVES To investigate the effects of lipid dysregulation on OA progression and to explore lipid dysregulation-targeting OA treatment of metformin. METHODS RNA-Seq data, biochemical, and histochemical assays in human and murine OA cartilage as well as primary chondrocytes were utilized to determine lipid dysregulation. Effects of metformin, a potent lipid-lowering medication, on ACSL4 expression and chondrocyte metabolism were determined. Further molecular experiments, including RT-qPCR, western blotting, flow cytometry, and immunofluorescence staining, were performed to investigate underlying mechanisms. Mice with intra-articular injection of metformin were utilized to determine the effects on ACLT-induced OA progression. RESULTS ACSL4 and 4-HNE expressions were elevated in human and ACLT-induced mouse OA cartilage and IL-1β-treated chondrocytes (P < 0.05). Ferrostatin-1 largely rescued IL-1β-induced MDA, lipid peroxidation, and ferroptotic mitochondrial morphology (P < 0.05). Metformin decreased the levels of OA-related genes (P < 0.05) and increased the levels of p-AMPK and p-ACC in IL-1β-treated chondrocytes. Intra-articular injection of metformin alleviated ACLT-induced OA lesions in mice, and reverted the percentage of chondrocytes positive for MMP13, Col2a1, ACSL4 and 4-HNE in ACLT mice (P < 0.05). Ferroptotic chondrocytes promoted the recruitment and chemotaxis of RAW264.7 cells via CCL2, which was blocked by metformin in vitro (P < 0.05). CONCLUSION We establish a critical role of polyunsaturated fatty acids metabolic process in OA cartilage degradation and define metformin as a potential OA treatment. Metformin reshapes lipid availability and ameliorates chondrocyte ferroptosis sensitivity via the AMPK/ACC pathway. In the future, gene-edited animals and extensive omics technologies will be utilized to reveal detailed lipids' involvement in cartilage lesions.
Collapse
Affiliation(s)
- Zhi Zou
- College of Bioengineering, Chongqing University, Chongqing 400044, China; Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Kang
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhonghua Xu
- Joint Disease & Sport Medicine Center, Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yuheng Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing Zhang
- College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jianmei Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yuan Zhang
- Joint Disease & Sport Medicine Center, Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| | - Shiwu Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
38
|
Ryan M, Megyeri S, Nuffer W, Trujillo JM. The potential role of GLP-1 receptor agonists in osteoarthritis. Pharmacotherapy 2025; 45:177-186. [PMID: 39980227 DOI: 10.1002/phar.70005] [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: 11/16/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 02/22/2025]
Abstract
Osteoarthritis (OA) is the most common form of arthritis, affecting over 500 million people globally. Current treatments are primarily symptom-focused, with no approved therapies to halt disease progression. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), widely used in type 2 diabetes (T2D) and obesity, demonstrate significant weight loss and glucose-lowering effects and have been shown to possess anti-inflammatory properties. Given the central role of inflammation and metabolic dysfunction in OA, this review examines the potential utility of GLP-1 RAs in OA management, focusing on both indirect effects, such as weight reduction, and possible direct effects on inflammatory pathways and cartilage preservation. Clinical studies suggest that GLP-1 RAs may benefit people with OA by reducing weight, improving glycemic control, and modulating inflammatory markers relevant to OA progression. Notable findings include significant weight loss and pain reduction in people with knee OA (KOA) treated with semaglutide in the STEP-9 trial. In other studies, GLP-1 RAs have shown potential to lower oxidative stress and pro-inflammatory cytokines, such as tumor necrosis factor (TNF-α) and interleukin (IL)-6, with reductions in OA-related pain and functional impairment observed in some cohorts. However, results vary, with some studies showing limited effects, potentially linked to the degree of weight loss achieved. Although some studies report variability in pain relief, likely influenced by the degree of weight loss achieved, GLP-1 RAs have shown overall promise in reducing both OA symptoms and markers associated with disease progression. This emerging evidence supports the utility of GLP-1 RAs as a potential disease-modifying option for OA, offering a dual benefit in metabolic and joint health. Future research should focus on establishing the long-term efficacy and safety and elucidating the mechanism by which GLP-1 RAs influence OA pathology.
Collapse
Affiliation(s)
- Mackenzie Ryan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Saige Megyeri
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Wes Nuffer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Jennifer M Trujillo
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
39
|
Anderson JR, Phelan MM, Caamaño-Gutiérrez E, Clegg PD, Rubio-Martinez LM, Peffers MJ. Metabolomic and proteomic stratification of equine osteoarthritis. Equine Vet J 2025. [PMID: 39972657 DOI: 10.1111/evj.14490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/14/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Equine osteoarthritis (OA) is predominantly diagnosed through clinical examination and radiography, leading to detection only after significant joint pathology. The pathogenesis of OA remains unclear and while many medications modify the disease's inflammatory components, no curative or reversal treatments exist. Identifying differentially abundant metabolites and proteins correlated with osteoarthritis severity could improve early diagnosis, track disease progression, and evaluate responses to interventions. OBJECTIVES To identify molecular markers of osteoarthritis severity based on histological and macroscopic grading. STUDY DESIGN Cross-sectional study. METHODS Post-mortem synovial fluid was collected from 58 Thoroughbred racehorse joints and 83 joints from mixed breeds. Joints were histologically and macroscopically scored and categorised by OA and synovitis grade. Synovial fluid nuclear magnetic resonance metabolomic and mass spectrometry proteomic analyses were performed, individually and combined. RESULTS In Thoroughbreds, synovial fluid concentrations of metabolites 2-aminobutyrate, alanine and creatine were elevated for higher OA grades, while glutamate was reduced for both Thoroughbreds and mixed breeds. In mixed breeds, concentrations of three uncharacterised proteins, lipopolysaccharide binding protein and immunoglobulin kappa constant were lower for higher OA grades; concentrations of an uncharacterised protein were higher for OA grade 1 only, and apolipoprotein A1 concentrations were higher for OA grades 1 and 2 compared with lower grades. For Thoroughbreds, gelsolin concentrations were lower for higher OA grades, and afamin was lower at a higher synovitis grade. Correlation analyses of combined metabolomics and proteomics datasets revealed 58 and 32 significant variables for Thoroughbreds and mixed breeds, respectively, with correlations from -0.48 to 0.42 and -0.44 to 0.49. MAIN LIMITATIONS The study's reliance on post-mortem assessments limits correlation with clinical osteoarthritis severity. CONCLUSIONS Following stratification of equine OA severity through histological and macroscopic grading, synovial fluid metabolomic and proteomic profiling identified markers that may support earlier diagnosis and progression tracking. Further research is needed to correlate these markers with clinical osteoarthritis severity.
Collapse
Affiliation(s)
- James R Anderson
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Veterinary Anatomy, Physiology and Pathology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Marie M Phelan
- NMR Metabolomics Facility, Liverpool Shared Research Facilities (LivSRF) & Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| | - Eva Caamaño-Gutiérrez
- Computational Biology Facility, Technology Directorate & Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| | - Peter D Clegg
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Luis M Rubio-Martinez
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Equine Clinical Science, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK
- Sussex Equine Hospital, West Sussex, UK
| | - Mandy J Peffers
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
40
|
Yu F, Zhu C, Wu W. Senile Osteoarthritis Regulated by the Gut Microbiota: From Mechanisms to Treatments. Int J Mol Sci 2025; 26:1505. [PMID: 40003971 PMCID: PMC11855920 DOI: 10.3390/ijms26041505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/19/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive degenerative joint disease that affects the entire synovial joint, leading to the progressive degeneration of articular cartilage. It seriously affects the quality of life and global disability of patients. OA is affected by a variety of factors; the most significant risk factor for OA is age. As individuals age, the risk and severity of OA increase due to the exacerbation of cartilage degeneration and wear and tear. In recent years, research has indicated that the gut microbiota may play a significant role in the aging and OA processes. It is anticipated that regulating the gut microbiota may offer novel approaches to the treatment of OA. The objective of this paper is to examine the relationship between the gut microbiota and senile OA, to investigate the potential mechanisms involved. This review also summarizes the therapeutic strategies related to gut flora in OA management, such as prebiotics and probiotics, diet, exercise, traditional Chinese medicine (TCM) modification, and fecal microbiota transplantation (FMT), highlighting the potential clinical value of gut flora and elucidating the current challenges. The foundation for future research directions is established through the summarization of current research progress.
Collapse
Affiliation(s)
- Fan Yu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (F.Y.); (C.Z.)
| | - Chenyu Zhu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (F.Y.); (C.Z.)
| | - Wei Wu
- School of Athletic Performance, Shanghai University of Sports, Shanghai 200438, China
| |
Collapse
|
41
|
Khan NM, Kaiser JM, Chihab S, Eng T, Drissi H. Dimethyl Fumarate attenuates synovial inflammation, reduces nociception, and inhibits the development of post-traumatic osteoarthritis. Biomed Pharmacother 2025; 183:117865. [PMID: 39862703 DOI: 10.1016/j.biopha.2025.117865] [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: 11/02/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025] Open
Abstract
There is currently no cure or disease-modifying treatment for post-traumatic osteoarthritis (PTOA). This study aims to assess the efficacy of dimethyl fumarate (DMF), a US-FDA approved drug for multiple sclerosis, as a treatment for PTOA. PTOA was induced in male Lewis rats by medial meniscal transection (MMT) surgery, and DMF was intra-articularly administered once, one week following surgery. PTOA progression was evaluated using histological, molecular, and radiographic analyses, while secondary allodynia was measured longitudinally, and pain-related markers expression were analyzed in dorsal root ganglion (DRG). 3D radiographic imaging by µCT analysis revealed that DMF treatment attenuated cartilage degradation by decreasing cartilage lesions, surface roughness, and osteophyte formation in the proximal tibiae. Histological analysis showed that DMF markedly inhibited cartilage erosion and cartilage surface fibrillation. Gene expression and Luminex analysis indicated that DMF suppressed joint inflammation by inhibiting inflammatory cytokines. DMF mitigated allodynic pain behavior at 6 weeks and repressed pain mediator expression (Calca, Tac1, Trpv1) in lumbar DRGs. Additionally, DMF treatment inhibited inflammatory gene expression and cytokine secretion induced by IL1β stimulation of human articular chondrocytes in vitro. Mechanistically, DMF treatment reduced colony-stimulating factor 2 (CSF2 or GM-CSF) level in the synovial fluid in vivo and inhibited its expression in human OA chondrocytes. Furthermore, siRNA targeting of CSF2 reduced inflammatory gene expression in human chondrocytes. The findings suggest that DMF reduced inflammatory gene expression, inhibited cartilage degeneration, and mitigated PTOA development in rats. It also alleviated pain behavior indicating its potential as a disease-modifying therapy for PTOA.
Collapse
Affiliation(s)
- Nazir M Khan
- Joseph Maxwell Cleland Atlanta VA Medical Center, Decatur, GA 30033, USA; Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA.
| | - Jarred M Kaiser
- Joseph Maxwell Cleland Atlanta VA Medical Center, Decatur, GA 30033, USA; Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA
| | - Samir Chihab
- Joseph Maxwell Cleland Atlanta VA Medical Center, Decatur, GA 30033, USA; Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA
| | - Tracy Eng
- Joseph Maxwell Cleland Atlanta VA Medical Center, Decatur, GA 30033, USA; Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA
| | - Hicham Drissi
- Joseph Maxwell Cleland Atlanta VA Medical Center, Decatur, GA 30033, USA; Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA.
| |
Collapse
|
42
|
Jiang N, Yang S, Sun Y, Zhang C, Liu K, Huang Y, Li F. The effect of exosomes from canine bone mesenchymal stem cells on IL-1β-mediated inflammatory responses in chondrocytes. Cytotechnology 2025; 77:27. [PMID: 39736844 PMCID: PMC11682030 DOI: 10.1007/s10616-024-00685-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 12/16/2024] [Indexed: 01/01/2025] Open
Abstract
Osteoarthritis is a degenerative disease of cartilage, and exosome derived from mesenchymal stem cells (MSCs) are considered promising for treating inflammatory musculoskeletal disorders, although their mechanisms are not fully understood. This study aimed to investigate the effects of exosomes derived from canine bone marrow mesenchymal stem cells (cBMSCs-Exos) on the expression of inflammatory factors and genes related cartilage matrix metabolism in IL-1β-induced canine chondrocytes. Canine BMSCs were isolated and characterized for surface markers and trilineage differentiation. Exosomes were then extracted and performed surface labeling detection. Canine chondrocytes were exposed to IL-1β to mimic osteoarthritis in vitro. Subsequently, the chondrocytes were treated with exosomes from BMSCs, and the expression levels of related genes and IL-6 protein were assessed. The mesenchymal stem cells isolated from bone marrow and cultured exhibited positive CD44 and CD90, negative expression of CD45 and HLA, and demonstrated potential to differentiate into adipocytes, osteoblasts and chondrocytes. Exosomes from BMSCs exhibited positivity expression of CD9, CD63 and CD81. Treatment with exosomes significantly reduced IL-6 and TNF-α mRNA levels induced by IL-1β, as well as IL-6 protein expression. Additionally, a significant decrease was observed in the mRNA levels catabolic marker genes MMP-13, ADAMTS-5, and COX2. Conversely, there was a significant increase in the mRNA levels of anti-inflammatory cytokines IL-4, IL-10, and anabolic marker genes, such as COL2A1, ACAN, and SOX9. cBMSCs-Exos play a vital role in cartilage protection by suppressing the expression of pro-inflammatory and anabolic genes while simultaneously enhancing the expression of genes involved in synthesis metabolism.
Collapse
Affiliation(s)
- Nan Jiang
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| | - Shuna Yang
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| | - Yunfei Sun
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| | - Chao Zhang
- Scholl of Biotechnology, Jiuquan Vocational Technical College, Jiuquan, 735000 China
| | - Kaicheng Liu
- Qingdao Kangdi’en Animal Pharmaceutical Co., Ltd, Qingdao, 266041 China
| | - Yufeng Huang
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| | - Fangzheng Li
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| |
Collapse
|
43
|
Walters M, Skovgaard K, Heegaard PMH, Fang Y, Kharaz YA, Bundgaard L, Skovgaard LT, Jensen HE, Andersen PH, Peffers MJ, Jacobsen S. Identification and characterisation of temporal abundance of microRNAs in synovial fluid from an experimental equine model of osteoarthritis. Equine Vet J 2025. [PMID: 39775906 DOI: 10.1111/evj.14456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND MicroRNAs, a class of small noncoding RNAs, serve as post-transcriptional regulators of gene expression and are present in a stable and quantifiable form in biological fluids. MicroRNAs may influence intra-articular responses and the course of disease, but very little is known about their temporal changes in osteoarthritis. OBJECTIVES To identify miRNAs and characterise the temporal changes in their abundance in SF from horses with experimentally induced osteoarthritis. We hypothesised that the abundance of miRNA would change during disease progression. STUDY DESIGN In vivo experiments. METHODS RNA extracted from synovial fluid obtained sequentially (Day 0, 28 and 70) from nine horses with experimentally induced osteoarthritis was subjected to small RNA sequencing using the Illumina Hiseq 4000 sequencing platform. Differentially abundant miRNAs underwent further validation and mapping of temporal abundance (Day 0, 14, 17, 21, 28, 35, 42, 49, 56, 63 and 70 days after osteoarthritis induction) by microfluidic reverse transcription quantitative real-time PCR. Bioinformatic analyses were performed to predict potential biological associations and target genes of the differentially abundant microRNAs. RESULTS Small RNA sequencing revealed 61 differentially abundant microRNAs at an early osteoarthritis stage (Day 28), and subsequent reverse transcription quantitative real-time PCR analysis validated 20 of these. Significant biological functions of the differentially abundant microRNAs were apoptosis, necrosis, cell proliferation and cell invasion. Following validation, four microRNAs (miRNA-199b-3p, miRNA-139-5p, miRNA-1839 and miRNA-151-5p) were detected in more than 50% of the synovial fluid samples and had higher abundance in osteoarthritic than in control joints. MAIN LIMITATIONS Limited sample size. CONCLUSION This is the first study to determine longitudinal changes in synovial fluid microRNA abundance in an equine model of osteoarthritis. Larger studies are needed in naturally occurring osteoarthritis to interrogate putative changes identified by this study.
Collapse
Affiliation(s)
- Marie Walters
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Peter M H Heegaard
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Yongxiang Fang
- Centre for Genomic Research, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Yalda A Kharaz
- Department of Musculoskeletal Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Louise Bundgaard
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Lene T Skovgaard
- Department of Public Health, University of Copenhagen, Copenhagen K, Denmark
| | - Henrik E Jensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Pia H Andersen
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Mandy J Peffers
- Department of Musculoskeletal Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| |
Collapse
|
44
|
Lu W, Feng W, Zhen H, Jiang S, Li Y, Liu S, Ru Q, Xiao W. Unlocking the therapeutic potential of WISP-1: A comprehensive exploration of its role in age-related musculoskeletal disorders. Int Immunopharmacol 2025; 145:113791. [PMID: 39667044 DOI: 10.1016/j.intimp.2024.113791] [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: 05/15/2024] [Revised: 10/03/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024]
Abstract
As the global population ages, the incidence of age-related musculoskeletal diseases continues to increase, driven by numerous complex and poorly understood factors. WNT-1 inducible secreted protein 1 (WISP-1), a secreted matrix protein, plays a critical role in the growth and development of the musculoskeletal system, including chondrogenesis, osteogenesis, and myogenesis. Numerous in vivo and in vitro studies have demonstrated that WISP-1 is significantly upregulated in age-related musculoskeletal conditions, such as osteoarthritis, osteoporosis, and sarcopenia, suggesting its involvement in the pathogenesis of these diseases. Regulating WISP-1 expression holds promise as a therapeutic strategy for improving musculoskeletal function, potentially offering new avenues for treating age-related musculoskeletal diseases in clinical practice. This review highlights the signaling pathways associated with WISP-1, its physiological roles within the musculoskeletal system, and its therapeutic potential in treating age-related musculoskeletal disorders.
Collapse
Affiliation(s)
- Wenhao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wenjie Feng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Haozu Zhen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Shide Jiang
- The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuguang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710001, Shaanxi, China.
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
45
|
Zheng H, Qu L, Yang L, Xie X, Song L, Xie Q. An injectable hydrogel loaded with Icariin attenuates cartilage damage in rabbit knee osteoarthritis via Wnt/β-catenin signaling pathway. Int Immunopharmacol 2025; 145:113725. [PMID: 39667046 DOI: 10.1016/j.intimp.2024.113725] [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: 07/30/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024]
Abstract
Knee osteoarthritis (KOA) is a chronic disease characterized by joint wear and cartilage degeneration. Current clinical treatments are based on symptomatic relief and are not effective in regenerating cartilage, and inflammation-induced cartilage damage accelerates the progression of osteoarthritis, making the protection of articular cartilage important for controlling the development of knee osteoarthritis. In this study, a biodegradable hydrogel (HA-Ca-Alg@Ica) loaded with Icariin (Ica) was prepared by in situ cross-linking of hyaluronic acid-calcium complex (HA-Ca) and sodium alginate (Alg-Na) for local sustained delivery of Ica. The hydrogel promoted chondrocyte proliferation and inhibited the degradation of cartilage matrix by regulating key factors (Wnt3a, β-catenin and GSK-3β) in the Wnt/β-catenin signaling pathway. In addition, the hydrogel reduced the expression of inflammatory factors, including IL-1β, IL-6, TNF-α, COX-2, and MMP13, leading to a reduction in inflammation and pain relief. In summary, this hydrogel containing Icariin has shown significant effects in reducing chondrocyte degradation and promoting chondrocyte proliferation, which can play a role in delaying osteoarthritis by protecting chondrocytes. These findings offer innovative prospects for the therapeutic management of knee osteoarthritis.
Collapse
Affiliation(s)
- Hanxiao Zheng
- The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Limin Qu
- College of Materials Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Lei Yang
- The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Xianmin Xie
- The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Ling Song
- College of Materials Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Qiuen Xie
- The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China.
| |
Collapse
|
46
|
Wei K, Zhou C, Shu Z, Shang X, Zou Y, Zhou W, Xu H, Liang Y, Ma T, Sun X, Xiao J. MYSM1 attenuates osteoarthritis by recruiting PP2A to deubiquitinate and dephosphorylate RIPK2. Bone Res 2025; 13:3. [PMID: 39746943 PMCID: PMC11696715 DOI: 10.1038/s41413-024-00368-y] [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: 12/05/2023] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 01/04/2025] Open
Abstract
Osteoarthritis (OA), the most prevalent degenerative joint disease, is marked by cartilage degradation and pathological alterations in surrounding tissues. Currently, no effective disease-modifying treatments exist. This study aimed to elucidate the critical roles of Myb-like, SWIRM, and MPN domains 1 (MYSM1) and its downstream effector, Receptor-interacting protein kinase 2 (RIPK2), in OA pathogenesis and the underlying mechanisms. Our findings revealed reduced MYSM1 levels in the cartilage of OA patients and mouse models. Genetic or adenovirus-induced MYSM1 knockout exacerbated OA progression in mice, whereas MYSM1 overexpression mitigated it. Mechanistically, MYSM1 inhibited the NF-κB and MAPK signaling pathways. Conversely, downstream RIPK2 significantly increased OA-like phenotypes and activated the NF-κB and MAPK pathways. The Ripk2S176D mutation accelerated OA pathogenesis, while Ripk2 silencing or Ripk2S176A mutation deactivated NF-κB and MAPK pathways, counteracting the role of MYSM1. MYSM1 deubiquitinates and dephosphorylates RIPK2S176 by recruiting protein phosphatase 2 A (PP2A). These results suggest that targeting MYSM1 or downstream RIPK2 offers promising therapeutic potential for OA.
Collapse
Affiliation(s)
- Kang Wei
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chuankun Zhou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zixing Shu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xingru Shang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Zou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Zhou
- Institute of Hepatobiliary Diseases, Transplant Center, Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Huanhuan Xu
- Department of Obstetrics and Gynecology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yulin Liang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuying Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
47
|
Kumavat R, Kumar V, Biswas S. Differential Expression of Fibrinogen Alpha and Its Potential Involvement in Osteoarthritis Pathogenesis. Mol Biotechnol 2025; 67:104-114. [PMID: 38182865 DOI: 10.1007/s12033-023-00983-w] [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: 08/11/2023] [Accepted: 11/05/2023] [Indexed: 01/07/2024]
Abstract
The deterioration of cartilage tissue and other joint components composed of synovial tissue is a defining characteristic of osteoarthritis (OA) disease. Because of the lack of understanding of the underlying cause and important molecular pathways, there are currently no effective diagnostic or treatment methods for OA. The purpose of the study is to find a specific protein biomarker with high sensitivity and specificity in order to understand the pathophysiology of the disease and the underlying molecular pathways. We examined plasma samples of matched age and sex from OA patients (n = 150) and healthy controls (HC) (n = 70) to find proteins that were differentially expressed and validated by western blotting, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and immunofluorescence. The results of western blotting demonstrated that the expression level of the fibrinogen alpha (FGA) protein was higher in plasma samples of osteoarthritis (OAPL) (p = 0.0343), and the ROC (receiver operating characteristic curve) curve supported the high sensitivity (95.22%) and specificity (74%) of FGA in OA plasma compared to healthy controls. FGA protein was detected to be deposited in the synovial tissue of OA patients (p = 0.0073). By activating the Toll-like receptor (TLR-4) receptor pathway in PBMCs (p = 0.04) and synovial tissue, FGA protein may be involved in the molecular mechanism of OA pathogenesis. Our findings collectively suggested that FGA, which is significantly expressed in OA plasma, synovial tissue, and PBMCs and is connected to the disease's advancement through the TLR-4 receptor, may serve as a diagnostic or disease-evolving tool for OA.
Collapse
Affiliation(s)
- Rajkamal Kumavat
- Council of Scientific &Industrial Research (CSIR) - Institute of Genomics & Integrative Biology, Mall Road, Delhi University Campus, 110007, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vijay Kumar
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sagarika Biswas
- Council of Scientific &Industrial Research (CSIR) - Institute of Genomics & Integrative Biology, Mall Road, Delhi University Campus, 110007, Delhi, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
- Department of Genomics & Molecular Medicine, Institute of Genomics and Integrative Biology, New Delhi, 110007, India.
| |
Collapse
|
48
|
Tassinari E, Minerba A, Basile T, Bucciarelli A, Traina F, Grigolo B, Zaffagnini S, Olivotto E. Post-operative injection of hydrolyzed collagen peptides shows anti-inflammatory effect in patients with femoroacetabular impingement improving the early recovery. J Exp Orthop 2025; 12:e70158. [PMID: 39896095 PMCID: PMC11783230 DOI: 10.1002/jeo2.70158] [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: 11/07/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 02/04/2025] Open
Abstract
Purpose This study aimed to compare the use of cortisone (C), intra-articular injected at the end of hip arthroscopy in patients with femoroacetabular impingement (FAI), to a new Class III medical device based on hydrolyzed collagen peptides 'PEPTYS' (P) and, to investigate potential associations among preoperative symptoms and hip function, outcomes after arthroscopic surgery and presence of inflammatory biomarkers in synovial fluids (SFs) at basal condition. Methods The two treatments were administrated to patients scheduled for arthroscopy with simple blind randomization sampling. Based on the sample size calculation, the number necessary to recruit was at least 20 patients for the C group and 20 for the P group. SFs, when available, were obtained by aspiration just prior to surgical intervention. At the baseline, osteoarthritis (OA) severity was assessed with a radiographic scoring system (Tönnis classification). Physical examination and clinical assessment using the Hip disability and Osteoarthritis Outcome Score (HOOS) and visual analogue scale (VAS) score for pain were performed at the time of surgery and at 1 and 6 months of follow-up. At the time of surgery, chondral (Outerbridge score) and labral pathology based on direct arthroscopic visualization were also evaluated. Results Forty-seven FAI patients were enroled, with a median age of 35 years with a standard deviation (SD) of 10.6 and a body mass index of 24.3kg/m² with an SD of 4.5. 24 patients were treated with C and 23 with P. Both treatments did not show any statistically significant difference in hip function and pain. High expression of inflammatory molecules in SFs was correlated with the worst post-operative articular function. Conclusions Our study showed that the use of P was completely comparable to cortisone. Therefore, PEPTYS might be a valuable candidate to improve early recovery, in terms of pain and function, from arthroscopic FAI treatment. Level of Evidence Level III, comparative and randomized study.
Collapse
Affiliation(s)
- Enrico Tassinari
- 2nd Orthopaedic and Traumatologic ClinicIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Andrea Minerba
- Orthopaedic‐Traumatology and Prosthetic Surgery and Revisions of Hip and KneeIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Tommaso Basile
- Orthopaedic‐Traumatology and Prosthetic Surgery and Revisions of Hip and KneeIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Alessio Bucciarelli
- RAMSES Laboratory, RIT Department, Research Centre Codivilla‐PuttiIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Francesco Traina
- Orthopaedic‐Traumatology and Prosthetic Surgery and Revisions of Hip and KneeIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Brunella Grigolo
- RAMSES Laboratory, RIT Department, Research Centre Codivilla‐PuttiIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Stefano Zaffagnini
- 2nd Orthopaedic and Traumatologic ClinicIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Eleonora Olivotto
- RAMSES Laboratory, RIT Department, Research Centre Codivilla‐PuttiIRCCS Istituto Ortopedico RizzoliBolognaItaly
| |
Collapse
|
49
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [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: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
50
|
Takemoto N, Takata Y, Shima Y, Goshima K, Shimozaki K, Kimura M, Kanayama T, Demura S, Nakase J. Blood flow signals through the bone cortex on ultrasonography can be used as a screening test for detecting bone marrow lesions on magnetic resonance imaging in patients with early knee osteoarthritis. J Med Ultrason (2001) 2025; 52:149-155. [PMID: 39365403 DOI: 10.1007/s10396-024-01503-2] [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: 05/21/2024] [Accepted: 09/01/2024] [Indexed: 10/05/2024]
Abstract
PURPOSE Blood flow signals (BFSs) through the bone cortex on ultrasonography (US) and bone marrow lesions (BMLs) detected on magnetic resonance imaging (MRI) can be used to assess bone lesions; however, no studies have reported their relationship. Therefore, this study aimed to assess whether BFSs through the bone cortex on US can serve as a screening test for detecting BMLs on MRI in patients with early knee osteoarthritis (OA). METHODS This study enrolled patients with knee joint pain who were diagnosed with early knee OA between January 2018 and January 2024. We targeted 77 patients who underwent MRI and in whom the presence or absence of BFSs through the bone cortex was confirmed on US. The association between BFSs and BMLs was evaluated using the chi-square test, and the sensitivity and specificity of BFSs for detecting BMLs on MRI were calculated. RESULTS The chi-square test showed that BFSs and BMLs were significantly associated in the femur and tibia (femur: χ2 [1] = 52.9, p < 0.001; Tibia: χ2 [1] = 44.8, p < 0.001). The sensitivity, specificity, positive predictive value, and negative predictive value of BFSs for detecting BMLs on MRI were 85.0%, 96.5%, 89.5%, and 94.8%, respectively, for the femur, and 84.0%, 92.3%, 84.0%, and 92.3%, respectively, for the tibia. CONCLUSION BFSs through the bone cortex on US can be used as a screening test for detecting BMLs on MRI in patients with early knee OA.
Collapse
Affiliation(s)
- Naoki Takemoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan.
| | - Yasushi Takata
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Yosuke Shima
- Department of Orthopedic Surgery, KKR Hokuriku Hospital, Kanazawa, Ishikawa, 921-8035, Japan
| | - Kenichi Goshima
- Department of Orthopedic Surgery and Joint Reconstructive Surgery, Kanazawa Munehiro Hospital, Kanazawa, Ishikawa, 920-0923, Japan
| | - Kengo Shimozaki
- Department of Orthopedic Surgery, Nomi Municipal Hospital, Nomi, Ishikawa, 929-0122, Japan
| | - Mitsuhiro Kimura
- Department of Orthopedic Surgery, Fukui General Hospital, Fukui, 910-3113, Japan
| | - Tomoyuki Kanayama
- Department of Orthopedic Surgery, KKR Hokuriku Hospital, Kanazawa, Ishikawa, 921-8035, Japan
| | - Satoru Demura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Junsuke Nakase
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| |
Collapse
|