1
|
Reihs E, Fischer A, Gerner I, Windhager R, Toegel S, Zaucke F, Rothbauer M, Jenner F. Beyond symptomatic alignment: evaluating the integration of causal mechanisms in matching animal models with human pathotypes in osteoarthritis research. Arthritis Res Ther 2025; 27:109. [PMID: 40382623 PMCID: PMC12084918 DOI: 10.1186/s13075-025-03561-4] [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: 10/09/2024] [Accepted: 04/14/2025] [Indexed: 05/20/2025] Open
Abstract
Osteoarthritis (OA) is a highly prevalent and disabling condition lacking curative treatments, with only symptomatic relief available. Recognizing OA as a heterogenous disorder with diverse aetiologies and molecular foundations underscores the need to classify patients by both phenotypes and molecular pathomechanisms (endotypes). Such stratification could enable the development of targeted therapies to surmount existing treatment barriers. From a scientific, economic, and ethical perspective, it is crucial to employ animal models that accurately represent the endotype of the target patient population, not merely their clinical symptoms. These models must also account for intrinsic and extrinsic factors, like age, sex, metabolic status, and comorbidities, which impact OA's pathogenesis and its clinical and molecular variability and can profoundly influence not only structural and symptomatic disease severity and progression but also the underlying molecular pathophysiology. The molecular definition of the OA subpopulation must also be reflected in the read-outs, as the traditional methods-macroscopic and histological scoring, along with limited gene expression profiling of established biomarkers for cartilage degradation, extracellular matrix (ECM) turnover, and synovial inflammation-are inadequate for discovering new, phenotype- and endotype-specific biomarkers or therapeutic targets. Thus, animal model characterisation should evolve to include both clinically and pathophysiologically pertinent measures of disease progression and response to treatment. This review evaluates the utility and accuracy of current animal models in OA research, focusing on their capacity to replicate the disease's pathophysiological processes.
Collapse
Affiliation(s)
- Eva Reihs
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Währinger Gürtel 18 - 20, Vienna, 1090, Austria
- Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Getreidemarkt 9/163, 1060, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Anita Fischer
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Währinger Gürtel 18 - 20, Vienna, 1090, Austria
| | - Iris Gerner
- Veterinary Tissue Engineering and Regenerative Medicine Vienna (VETERM), Equine Surgery Unit, University of Veterinary Medicine Vienna, Vienna, Veterinärplatz 1, 1210, Austria
| | - Reinhard Windhager
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Währinger Gürtel 18 - 20, Vienna, 1090, Austria
- Division of Orthopedics, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, 1090, Austria
| | - Stefan Toegel
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Währinger Gürtel 18 - 20, Vienna, 1090, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim GmbH, Maienburgstr. 2, Frankfurt/Main, 60528, Germany
| | - Mario Rothbauer
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Währinger Gürtel 18 - 20, Vienna, 1090, Austria.
- Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Getreidemarkt 9/163, 1060, Austria.
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria.
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria.
| | - Florien Jenner
- Veterinary Tissue Engineering and Regenerative Medicine Vienna (VETERM), Equine Surgery Unit, University of Veterinary Medicine Vienna, Vienna, Veterinärplatz 1, 1210, Austria
| |
Collapse
|
2
|
Zhao Z, Geng P, An M, Zhao Y, Guo Z, Gao H, Zhu H, Li Z, Wei M, Li C. Days 7 to 14 May Represent an Optimal Window for Stem Cell-Based Treatment in a Rat Model of Anterior Cruciate Ligament Transection-Induced Posttraumatic Osteoarthritis. Am J Sports Med 2025:3635465251326499. [PMID: 40336188 DOI: 10.1177/03635465251326499] [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] [Indexed: 05/09/2025]
Abstract
BACKGROUND The concept of early intervention at the appropriate time has been recognized for treating posttraumatic osteoarthritis (PTOA). However, the optimal intervention timing to achieve superior therapeutic efficacy remains unclear. In addition, there is a lack of direct evidence regarding therapeutic efficacies achieved at different time points. HYPOTHESIS The administration schedule of stem/stromal cells can significantly influence their ability to improve function and slow progression of PTOA. There may exist an appropriate time window for achieving superior therapeutic efficacy. STUDY DESIGN Controlled laboratory study. METHODS A total of 72 Sprague Dawley rats were included in this study. Anterior cruciate ligament transection (ACLT) was performed to induce PTOA. Animals in the control group underwent ACLT alone, whereas those in the sham group underwent knee articular capsulotomy alone. Bone marrow mesenchymal stem/stromal cells or phosphate-buffered saline (PBS) was intra-articularly administered on days 3, 7, 14, and 28 after ACLT (n = 6). Bioluminescence imaging was used to detect the retention of stem cells administered at different time points (n = 3). At the end of the experiment (8 weeks), gait analysis was conducted using CatWalk to compare the recovery of knee function between the 2 groups. Micro-computed tomography (CT) was performed to assess general appearance and quantify the microstructure of subchondral bone. Histological staining was used to evaluate the whole-joint pathology. Semiquantitative evaluations were conducted using Osteoarthritis Research Society International and Mankin scores. RESULTS PBS administration at different time points had no therapeutic effects on lower limb function or PTOA progression. Gait analysis suggested that stem cell administration significantly improved the general function of knee joints compared with the control group at all time points. However, the duty cycle was significantly higher on days 7 and 14 after ACLT. Micro-CT and histopathological staining of the knee samples suggested that although stem cell administration significantly ameliorated the progression of PTOA, the therapeutic efficacy was significantly better on days 7 and 14. After stem cell administration, the articular surface was considerably smoother with few scattered osteophytes, the deposition of cartilage extracellular matrix was more abundant, subchondral bone remodeling was significantly alleviated, and the synovium was less hyperplastic with reduced inflammatory cell infiltration. The general retention time of stem cells did not differ significantly at different administration time points. CONCLUSION This study suggests that the intervention schedule is significantly correlated with the therapeutic efficacy of stem cells for PTOA, with the best effects observed on days 7 and 14 after ACLT. CLINICAL RELEVANCE Days 7 to 14 after trauma may be the appropriate intervention timing for clinical prevention and treatment of PTOA.
Collapse
Affiliation(s)
- Zhidong Zhao
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Peng Geng
- Department of Ultrasound, Aerospace Center Hospital, Beijing, China
| | - Mingyang An
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Yanpeng Zhao
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Zheng Guo
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Huayi Gao
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Heng Zhu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhongli Li
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Min Wei
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Chunbao Li
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
3
|
Chen M, Yang Y, Cui M, Wang B. Solute transport from synovial fluid to articular cartilage and subchondral bone at different stages of osteoarthritis in a live mouse model. J Orthop Translat 2025; 52:291-300. [PMID: 40421143 PMCID: PMC12104021 DOI: 10.1016/j.jot.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/19/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Objective This study aims to (1) identify a simplified method to preserve sample integrity and maintain original fluorescence distribution; (2) assess the diffusivity of small and large molecules within articular cartilage (AC), calcified cartilage (CC), and subchondral bone (SB); and (3) investigate the changes in solute transport at various stages of osteoarthritis (OA) in a destabilization of the medial meniscus (DMM) murine model. Methods Fluorescent dyes of small and large molecules were injected into the knee joints of live mice. Joints were harvested and rapidly frozen immediately post-euthanasia. Optimal dye concentrations and dwelling times were determined through exploratory studies. Mice underwent either DMM or sham surgery and were evaluated at 2 and 8 weeks postoperatively. Relative fluorescence intensity was quantified within the AC, CC and SB, complemented by micro-CT, safranin O staining, and collagen II immunohistochemistry staining. Results The methodology successfully preserved sample integrity and original dye distribution. Fluorescent imaging revealed that small solute was mainly restricted by the tidemark, while large solute showed limited permeability in AC. Permeability of AC remained elevated in the DMM group at both time points. Increased permeability in CC and SB was observed only at 8 weeks post-DMM surgery, accompanied by reduced collagen II amount. Conclusions In live mice, the tidemark serves as a barrier to small molecule diffusion, while the cartilage surface restricts larger molecules; however, both structures exhibit increased permeability in OA. These findings advance the understanding of OA pathogenesis and suggest potential therapeutic targets related to cartilage permeability. Translational Potential The findings of this study advance the understanding of osteoarthritis pathogenesis by elucidating the role of solute transport alterations in cartilage and subchondral bone, thereby suggesting potential therapeutic targets aimed at modulating cartilage permeability to improve joint health in osteoarthritis.
Collapse
Affiliation(s)
- Mengcun Chen
- Center for Translational Medicine, Departments of Medicine and Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| | - Yanmei Yang
- Center for Translational Medicine, Departments of Medicine and Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| | - Mingshu Cui
- Center for Translational Medicine, Departments of Medicine and Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| | - Bin Wang
- Center for Translational Medicine, Departments of Medicine and Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, United States
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| |
Collapse
|
4
|
Yang T, Yang X, Wang G, Jia D, Li Y. Unraveling the crucial role of SDF-1 in osteoarthritis progression: IL6/HIF-1α positive feedback and chondrocyte ferroptosis. Int Immunopharmacol 2025; 152:114400. [PMID: 40058106 DOI: 10.1016/j.intimp.2025.114400] [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: 01/07/2025] [Revised: 02/15/2025] [Accepted: 02/27/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND Osteoarthritis (OA) is a common joint disease with an incompletely understood pathogenesis. SDF-1, a key factor in cartilage matrix degradation, is involved in OA cartilage degeneration, yet its mechanism, especially regarding ferroptosis, remains unclear. This study focuses on elucidating the role of SDF-1-induced chondrocyte ferroptosis and the IL6/HIF-1α signalling axis in OA. METHODS A rabbit OA model was created via SDF-1 induction. Knee cartilage tissues were sequenced and analyzed bioinformatically to identify key genes, and explore critical pathways. Clinical tissue samples were utilized to validate their clinical relevance. Furthermore, cell and rabbit models were constructed through gene interference and pathway blocking. The expression of related genes and proteins was detected by QPCR, ELISA, Western blot, and immunofluorescence. Additionally, OA and ferroptosis indicators such as cell viability, immunohistochemistry, ROS, lipid ROS, Fe2+, MDA, and mitochondrial morphology were evaluated to uncover the molecular mechanism by which SDF-1 regulates the IL6/HIF-1α signalling axis to mediate chondrocyte ferroptosis. RESULTS Bioinformatics revealed that ferroptosis was significantly activated in SDF-1-induced OA, with IL6 and HIF-1 pathways implicated. In vitro and in vivo, SDF-1 increased the expression and secretion of MMP13 but decreased COL2A1 and ACAN in chondrocytes, leading to OA-like changes. It also suppressed the expression levels of SLC7A11 and GPX4, upregulated the gene and protein levels of ACSL4, promoted the accumulation of MDA, Fe2+, and ROS, and caused mitochondrial morphological changes. These ferroptosis manifestations could be alleviated by the ferroptosis inhibitor Fer-1. IL6 was an important mediator of SDF-1-induced ferroptosis, and knocking down IL6 also inhibited chondrocyte ferroptosis changes. Overexpressing IL6 (oeIL6) and using PX478 to inhibit the HIF-1 signalling pathway showed that PX478 could significantly relieve the cytotoxicity produced by the culture of oeIL6 and SDF-1, enhance chondrocyte viability, reverse the decreased expression of SLC7A11 and GPX4 caused by oeIL6, increase the expression of ACSL4, reverse the accumulation of MDA, Fe2+, and ROS. Moreover, PX478 could also significantly reduce the expression and secretion of IL6. CONCLUSION SDF-1 mediates chondrocyte ferroptosis via the IL6/HIF-1α positive feedback, promoting OA.
Collapse
Affiliation(s)
- Tengyun Yang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Xianguang Yang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Guoliang Wang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Di Jia
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Yanlin Li
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China.
| |
Collapse
|
5
|
Yogi A, Banderali U, Moreno MJ, Martina M. Preclinical Animal Models to Investigate the Role of Na v1.7 Ion Channels in Pain. Life (Basel) 2025; 15:640. [PMID: 40283194 PMCID: PMC12028925 DOI: 10.3390/life15040640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Chronic pain is a maladaptive neurological disease that remains a major global healthcare problem. Voltage-gated sodium channels (Navs) are major drivers of the excitability of sensory neurons, and the Nav subtype 1.7 (Nav1.7) has been shown to be critical for the transmission of pain-related signaling. This is highlighted by demonstrations that gain-of-function mutations in the Nav1.7 gene SCN9A result in various pain pathologies, whereas loss-of-function mutations cause complete insensitivity to pain. A substantial body of evidence demonstrates that chronic neuropathy and inflammation result in an upregulation of Nav1.7, suggesting that this channel contributes to pain transmission and sensation. As such, Nav1.7 is an attractive human-validated target for the treatment of pain. Nonetheless, a lack of subtype selectivity, insufficient efficacy, and adverse reactions are some of the issues that have hindered Nav1.7-targeted drug development. This review summarizes the pain behavior profiles mediated by Nav1.7 reported in multiple preclinical models, outlining the current knowledge of the biophysical, physiological, and distribution properties required for a Nav1.7 inhibitor to produce analgesia.
Collapse
Affiliation(s)
- Alvaro Yogi
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON K1A 0R6, Canada; (U.B.); (M.J.M.); (M.M.)
| | | | | | | |
Collapse
|
6
|
Mirazi H, Wood ST. Microfluidic chip-based co-culture system for modeling human joint inflammation in osteoarthritis research. Front Pharmacol 2025; 16:1579228. [PMID: 40271077 PMCID: PMC12015981 DOI: 10.3389/fphar.2025.1579228] [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: 02/18/2025] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Here we present a microfluidic model that allows for co-culture of human osteoblasts, chondrocytes, fibroblasts, and macrophages of both quiescent (M0) and pro-inflammatory (M1) phenotypes, maintaining initial viability of each cell type at 24 h of co-culture. We established healthy (M0-based) and diseased (M1-based) joint models within this system. An established disease model based on supplementation of IFN-γ and lipopolysaccharide in cell culture media was used to induce an M1 phenotype in macrophages to recapitulate inflammatory conditions found in Osteoarthritis. Cell viability was assessed using NucBlue™ Live and NucGreen™ Dead fluorescent stains, with mean viability of 83.9% ± 14% and 83.3% ± 12% for healthy and diseased models, respectively, compared with 93.3% ± 4% for cell in standard monoculture conditions. Cytotoxicity was assessed via a lactate dehydrogenase (LDH) assay and showed no measurable increase in lactate dehydrogenase release into the culture medium under co-culture conditions, indicating that neither model promotes a loss of cell membrane integrity due to cytotoxic effects. Cellular metabolic activity was assessed using a PrestoBlue™ assay and indicated increased cellular metabolic activity in co-culture, with levels 5.9 ± 3.2 times mean monolayer cell metabolic activity levels in the healthy joint model and 5.3 ± 3.4 times mean monolayer levels in the diseased model. Overall, these findings indicate that the multi-tissue nature of in vivo human joint conditions can be recapitulated by our microfluidic co-culture system at 24 h and thus this model serves as a promising tool for studying the pathophysiology of rheumatic diseases and testing potential therapeutics.
Collapse
Affiliation(s)
- Hosein Mirazi
- Department of Nanoscience and Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, SD, United States
| | - Scott T. Wood
- Department of Nanoscience and Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, SD, United States
- Portland Laboratory for Biotechnology and Health Sciences, University of New England, Portland, ME, United States
- Department of Biomedical Sciences, University of New England, Biddeford, ME, United States
| |
Collapse
|
7
|
Glinkowski W, Śladowski D, Tomaszewski W, Pol-IAHA Study Group. Molecular Mechanisms and Therapeutic Role of Intra-Articular Hyaluronic Acid in Osteoarthritis: A Precision Medicine Perspective. J Clin Med 2025; 14:2547. [PMID: 40283379 PMCID: PMC12027770 DOI: 10.3390/jcm14082547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/24/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Osteoarthritis (OA) is a degenerative joint disease characterized by progressive cartilage breakdown, synovial inflammation, and pain, which leads to significant disability. IAHA is widely used because of its viscoelastic properties, which restore synovial fluid homeostasis and reduce symptoms. However, emerging evidence suggests that IAHA exerts additional biological effects including chondroprotection, inflammatory modulation, oxidative stress reduction, and pain modulation, which may influence disease progression. Objective: This narrative review examines the biological mechanisms underlying IAHA's role in OA management. The review explored IAHA's effects on synovial fluid viscoelasticity, inflammatory cytokine modulation, cartilage preservation, oxidative stress regulation, and pain pathways, emphasizing the influence of molecular weight variations on therapeutic efficacy. Additionally, this review evaluates IAHA's integration into multimodal treatment strategies, its potential disease-modifying effects, and future directions for personalized treatment approaches. Methods: A comprehensive literature review was conducted using PubMed, Cochrane Library, EMBASE, Scopus, and Web of Science for studies published between January 2000 and March 2024. The search focused on IAHA's molecular, cellular, and biochemical effects in OA and clinical findings assessing its impact on joint function, pain relief, and disease progression. Results: IAHA improves synovial fluid lubrication, reduces proinflammatory cytokines (IL-1β, TNF-α), inhibits matrix metalloproteinases (MMPs), scavenges reactive oxygen species (ROS), and modulates nociceptive pathways. High-molecular-weight IAHA demonstrates superior efficacy in advanced OA, while low-molecular-weight formulations may be better suited for early-stage disease. Although IAHA's symptom relief is comparable to corticosteroids and NSAIDs, its favorable safety profile and emerging disease-modifying potential support its long-term use in OA management. Conclusions: IAHA represents a multifaceted therapeutic approach bridging symptomatic relief and regenerative strategies. While long-term efficacy, optimal administration protocols, and patient-specific responses remain subjects of ongoing research, refining treatment selection criteria, dosing regimens, and combination strategies may enhance clinical outcomes. Future studies should explore biomarker-driven approaches, standardize treatment protocols, and assess IAHA's synergy with regenerative medicine to optimize its role in OA management.
Collapse
Affiliation(s)
- Wojciech Glinkowski
- Center of Excellence “TeleOrto” for Telediagnostics and Treatment of Disorders and Injuries of the Locomotor System, Department of Medical Informatics and Telemedicine, Medical University of Warsaw, 02-091 Warsaw, Poland
- Stichting Med Partners, 1098 XH Amsterdam, The Netherlands
| | - Dariusz Śladowski
- Department of Transplantology and Central Tissue Bank, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Wiesław Tomaszewski
- ARS MEDICA Foundation for Medical Education, Health Promotion, Art and Culture, 03-301 Warsaw, Poland
- College of Physiotherapy, 50-038 Wrocław, Poland
| | | |
Collapse
|
8
|
Chen L, Yang J, Cai Z, Huang Y, Zhang Q, Zhou C, Wang J, Huang W, Cui W, Hu N. Cerium Dioxide Nanoparticles-Based Inspector Enhances Mitochondrial Quality Control to Maintain Chondrocyte Homeostasis in Osteoarthritis Therapy. Adv Healthc Mater 2025; 14:e2405069. [PMID: 40033885 DOI: 10.1002/adhm.202405069] [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: 12/15/2024] [Revised: 02/02/2025] [Indexed: 03/05/2025]
Abstract
Imbalanced mitochondrial quality control is strongly linked to the onset and development of osteoarthritis (OA). However, current research primarily focuses on local cartilage repair and phenotype maintenance, lacking a systematic approach to subcellular mitochondrial quality control. To address this, the present study proposes a mitochondrial quality control strategy based on nanozyme hydrogel microspheres ("mitochondrial inspector"), constructed through electrostatic self-assembly, incorporation of dynamic diselenide bonds, and microfluidic technology. The mitochondrial oxidative stress microenvironment is improved by cerium dioxide nanoparticles and combined with metformin to activate autophagy to clear persistently dysfunctional mitochondria, thereby inhibiting OA progression. In vitro results showed that "mitochondrial inspector" not only significantly improved the oxidative stress microenvironment of chondrocytes, but also efficiently scavenged the damaged mitochondria, increased the mitochondrial membrane potential by over 20-fold, and notably improved the mitochondrial function and chondrocyte homeostasis. In a rat OA model, minimally invasive intra-articular injection of the "mitochondrial inspector" effectively regulated mitochondrial quality, alleviated cartilage matrix degradation, reduced osteophyte formation by ≈80%, and reduced the Mankin score for cartilage damage by over 70%. In summary, this study presents a novel nanozyme microsphere-based mitochondrial quality control strategy for the treatment of OA, providing new insights for subcellular therapies for other aging-related diseases.
Collapse
Affiliation(s)
- Li Chen
- Department of Orthopedics, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jianye Yang
- Department of Orthopedics, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Zhengwei Cai
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yanran Huang
- Department of Orthopedics, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Qinyang Zhang
- Department of Orthopedics, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Chengqiang Zhou
- Department of Orthopedics, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Juan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wei Huang
- Department of Orthopedics, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Ning Hu
- Department of Orthopedics, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| |
Collapse
|
9
|
Glinkowski WM, Tomaszewski W. Intra-Articular Hyaluronic Acid for Knee Osteoarthritis: A Systematic Umbrella Review. J Clin Med 2025; 14:1272. [PMID: 40004802 PMCID: PMC11856182 DOI: 10.3390/jcm14041272] [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: 12/10/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Objective: to evaluate the efficacy, safety, and cost-effectiveness of intra-articular hyaluronic acid (IAHA) in treating osteoarthritis (OA), considering innovations in formulations, comparative outcomes, and variability in guidelines. This review aims to synthesize evidence supporting the role of IAHA in multimodal treatment strategies. Materials and Methods: A general, narrative, umbrella review of systematic reviews and meta-analyses was conducted. Clinical practice recommendations and guidelines for IAHA use were also reviewed and evaluated. A comprehensive search was conducted across the main medical data sources. Inclusion criteria focused on studies evaluating the efficacy, safety, and impact of IAHA. Key outcomes included pain reduction (e.g., WOMAC, VAS), functional improvement, safety, and cost-effectiveness. Results: IAHA showed moderate efficacy in pain relief and functional improvement, especially in early-to-moderate OA. The results indicate that hybrid formulations and combination therapies show better clinical outcomes, with expanded efficacy and potential chondroprotection. However, heterogeneity between studies was noted, reflecting variability in patient populations and intervention protocols. International guidelines varied significantly, with some opposing routine use (e.g., AAOS, NICE) and others endorsing IAHA more or less conditionally (e.g., ESCEO, OARSI). Conclusions: IAHA remains a treatment modality in the arsenal of selected populations of people with OA, especially for early and moderate disease. High-quality, standardized studies are still needed to refine IAHA's role and establish personalized guidelines for individual patients. A concerted effort to harmonize global recommendations and economic strategies, such as tiered pricing, can increase equitable access and optimize IAHA's integration of multimodal treatment for OA.
Collapse
Affiliation(s)
- Wojciech Michał Glinkowski
- Center of Excellence “TeleOrto” for Telediagnostics and Treatment of Disorders and Injuries of the Locomotor System, Department of Medical Informatics and Telemedicine, Medical University of Warsaw, 02-091 Warsaw, Poland
- Stichting Med Partners, 1098 XH Amsterdam, The Netherlands
| | - Wiesław Tomaszewski
- Ars Medica Foundation for Medical Education, Health Promotion, Art and Culture, 03-301 Warsaw, Poland
- College of Physiotherapy, 50-038 Wrocław, Poland
| |
Collapse
|
10
|
Zhu L, Bi Y, Liang T, Zhang P, Xiao X, Yu T. Ginkgetin delays the progression of osteoarthritis by inhibiting the NF-κB and MAPK signaling pathways. J Orthop Surg Res 2025; 20:139. [PMID: 39910626 PMCID: PMC11800635 DOI: 10.1186/s13018-025-05525-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA) is considered an advancing chronic degenerative joint disease, leading to severe physical functional impairment of patients. Its development is closely related to increased inflammation and oxidative stress within the joint. Ginkgetin (GK), a natural non-toxic chemical, has proven anti-inflammatory, antioxidant, anti-tumor, and neuroprotective effects. METHODS First, this study utilizes network pharmacology to explore the intrinsic connection between GK and OA. In vitro, SW1353 human cartilage cells were stimulated with Tert-butyl hydrogen peroxide (TBHP), and different GK concentrations were pre-treated to evaluate its protective effects. GK's anti-inflammatory and antioxidative effects were comprehensively assessed via MTT assay, western blot, cell immunofluorescence, ELISA, and transcriptome sequencing. Potential underlying mechanisms were also explored. In vivo, OA was induced in rats via anterior cruciate ligament transection (ACLT), and GK's impact on cartilage protection was further assessed via histological analysis and western blot. RESULTS Network pharmacology has revealed that GK regulates OA via several key pathways, especially NF-κB, HIF-1, PI3K-AKT, and substances like reactive oxygen species. In vitro experiments showed GK effectively reverses oxidative stress damage from TBHP, inhibits inflammatory factor release, and protects Extracellular matrix (ECM) from degradation. These functions may be achieved via the NF-κB and MAPK signaling pathways. In vivo experiments showed GK significantly reduced proteoglycan loss from ACLT and inhibited matrix metalloproteinase 13 (MMP13) and ADAMTS5 (A disintegrin and metalloproteinase with thrombospondin motifs 5) production, effectively preventing cartilage degeneration in rats. CONCLUSION These findings suggest that GK has potential as a therapeutic agent for OA, offering new strategies and directions for OA treatment.
Collapse
Affiliation(s)
- Liang Zhu
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yanchi Bi
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ting Liang
- Rehabilitation Section, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Po Zhang
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xiao Xiao
- Central Laboratories, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| | - Tengbo Yu
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
11
|
Chen Y, Zhang Y, Wu C, Zhao X, Zhang H, Li C, Deng Y, Sun L, Zhou Y, Zhang X. High-Throughput Screening Strategy and Metal-Organic Framework-Based Multifunctional Controlled-Release Nanomaterial for Osteoarthritis Therapy. ACS NANO 2025; 19:4802-4819. [PMID: 39829021 DOI: 10.1021/acsnano.4c15740] [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: 01/22/2025]
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease that lacks effective therapy. Oxidative stress is one of the major factors contributing to OA; however, treatments targeting oxidative stress are still lacking. In the current study, we established an oxidative stress-induced cell death model in chondrocytes in vitro and screened drugs that may suppress oxidative stress-induced cell death. Ethyl gallate (EG) was identified as the most potent drug against oxidative stress-induced cell death out of more than 600 drugs in the natural product library. Application of drugs without an appropriate delivery system for OA therapy may have drawbacks such as low bioavailability, short action time, and poor efficacy. Herein, poly-His6-zinc assembly (PZA), a pH-responsive metal-organic framework (MOF) loaded with EG (EG@PZA) was designed for OA therapy. It was demonstrated that EG@PZA may have the lysosome escape property, which dramatically increases the utilization of EG. Furthermore, EG@PZA showed enhanced release capability of EG in the acidic microenvironment. In vitro and in vivo studies demonstrated that EG@PZA effectively suppresses oxidative stress-induced extracellular matrix degradation, ferroptosis, and senescence in chondrocytes and also ameliorates OA in the destabilization of the medial meniscus (DMM) mouse model in vivo. Together, the current study showed that EG@PZA may become a potential controlled-release nanomaterial for effective OA therapy.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yekai Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chenyu Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaoying Zhao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hanwen Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chenchao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yuxin Deng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Liaojun Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
12
|
Xiao G, Qin J, Yang H, Song Q, Zhang R, Huang J, Mou Y, Liu W, Sun X, Nie M. Characterization of a chemically induced osteoarthritis model in zebrafish. Sci Rep 2025; 15:3905. [PMID: 39890962 PMCID: PMC11785742 DOI: 10.1038/s41598-025-88125-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 01/24/2025] [Indexed: 02/03/2025] Open
Abstract
Osteoarthritis (OA) is characterized by the progressive degeneration of the synovial joint, leading to irreversible damage to articular cartilage and subchondral bone. While animal models have advanced our understanding of OA, numerous unresolved issues still remain. The zebrafish, known for its transparent body, rapid developmental, and impressive regenerative capabilities, offers substantial potential for osteoarthritis research. This study seeks to establish a new OA model utilizing the zebrafish jaw joint, acting as a supplement to traditional animal models. In the future, this model could serve as a valuable platform for delving deeper into the mechanisms of this disease, as well as for advancing drug discovery and therapeutic interventions. Leveraging the skeletal structure of zebrafish, we targeted the largest jaw joint for our research. A custom fixation device was crafted, and a microinjection system was utilized to inject mono-iodoacetate (MIA) or collagenase type II (CTII) into the joint cavity of zebrafish. Subsequent analyses included histological staining, immunohistochemistry, OA research society international (OARSI) scoring, and real-time in vivo imaging were performed at 7, 14, and 28 days post injection. Our results effectively demonstrated the presence of synovial inflammation and cartilage damage within the zebrafish mandible, affirming the feasibility of inducing OA in zebrafish. In conclusion, the local injection of chemical agents into the joint cavity of zebrafish effectively induced the occurrence of OA. Establishing the zebrafish OA model enhances the array of animal models available for OA research. Moreover, zebrafish present distinct advantages, including robust regenerative abilities, genetic editing simplicity, and efficient drug screening. Consequently, this offers a fresh avenue for investigating the pathogenesis, prevention, and potential therapeutic approaches for human OA.
Collapse
Affiliation(s)
- Gongyi Xiao
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400010, China
- Department of Orthopedic Surgery, Chonggang General Hospital, No. 1 Dayan Sancun, Dadukou District, Chongqing, 400000, China
| | - Jin Qin
- Center for Spinal Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400010, China
| | - Huiping Yang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400010, China
| | - Qizhi Song
- Department of Orthopedic Surgery, Chonggang General Hospital, No. 1 Dayan Sancun, Dadukou District, Chongqing, 400000, China
| | - Ruobin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Junlan Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yuexi Mou
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400010, China
| | - Wen Liu
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400010, China
| | - Xianding Sun
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400010, China.
| | - Mao Nie
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400010, China.
| |
Collapse
|
13
|
Lei J, Chen X, Xie H, Dai Y, Chen Z, Xu L. Therapeutic efficacy of intra-articular injection of human adipose-derived mesenchymal stem cells in a sheep model of knee osteoarthritis. Stem Cell Res Ther 2025; 16:24. [PMID: 39849597 PMCID: PMC11755983 DOI: 10.1186/s13287-025-04143-6] [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: 11/06/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Mesenchymal stem cells have great potential for repairing articular cartilage and treating knee osteoarthritis (KOA). Nonetheless, little is known about the efficacy of human adipose-derived mesenchymal stem cells (haMSCs) for KOA in large animal models. METHODS This study evaluated the therapeutic efficacy of haMSCs in knee articular cartilage repair in a sheep model of KOA. haMSCs were isolated, cultured, and characterized. KOA was surgically induced by anterior cruciate ligament transection and medial meniscectomy, followed by intra-articular injection of saline (negative control group) or haMSCs (haMSC group) into the right knee joint at 6 and 9 weeks after surgery. Sheep were sacrificed 21 weeks after surgery, and samples (whole knee joints, femoral condyles, and tibias) were collected, processed, and analyzed. Changes in knee articular cartilage were assessed by magnetic resonance imaging, micro-computed tomography, macroscopic analysis, histology, and immunohistochemistry. RESULTS KOA caused the degeneration of the medial femoral condyle in the sheep model of KOA. Conversely, haMSCs repaired chondral defects and increased the thickness of knee articular cartilage. CONCLUSIONS These data suggest that the intra-articular injection of haMSCs can effectively repair articular cartilage defects in the knee.
Collapse
Affiliation(s)
- Jigang Lei
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Xingyi Chen
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Haohao Xie
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Yuhao Dai
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Zhongjin Chen
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Liang Xu
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China.
| |
Collapse
|
14
|
Griffin TM, Lopes EBP, Cortassa D, Batushansky A, Jeffries MA, Makosa D, Jopkiewicz A, Mehta-D'souza P, Komaravolu RK, Kinter MT. Sexually dimorphic metabolic effects of a high fat diet on knee osteoarthritis in mice. Biol Sex Differ 2024; 15:103. [PMID: 39639386 PMCID: PMC11619521 DOI: 10.1186/s13293-024-00680-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Women have a higher risk of developing osteoarthritis (OA) than men, including with obesity. To better understand this disparity, we investigated sex differences in metabolic and inflammatory factors associated with OA using a diet-induced mouse model of obesity. We hypothesized that 20 weeks of high-fat diet (HFD) would induce sexually dimorphic changes in both systemic and local risk factors of knee OA. METHODS Male and female C57BL/6J mice were fed Chow or HFD from 6 to 26 weeks of age (n = 12 per diet and sex). We performed broad metabolic phenotyping, 16 S gut microbiome analysis, targeted gene expression analysis of synovium-infrapatellar fat tissue, targeted gene expression and proteomic analysis of articular cartilage, chondrocyte metabolic profiling, and OA histopathology. Two-way ANOVA statistics were utilized to determine the contribution of sex and diet and their interaction on outcomes. RESULTS Mice fed HFD weighed 1.76-fold (p < 0.0001) and 1.60-fold (p < 0.0001) more than male and female Chow cohorts, respectively, with both sexes reaching similar body fat levels (male: 43.9 ± 2.2%; female: 44.1 ± 3.8%). HFD caused greater cartilage pathology (p < 0.024) and synovial hyperplasia (p < 0.038) versus Chow in both sexes. Cartilage pathology was greater in male versus female mice (p = 0.048), and only male mice developed osteophytes with HFD (p = 0.044). Both sexes exhibited metabolic inflexibility on HFD, but only male mice developed glucose intolerance (p < 0.0001), fatty liver (p < 0.0001), and elevated serum amylase (p < 0.0001) with HFD versus Chow. HFD treatment caused sex-dependent differences in gut microbiota beta diversity (p = 0.01) and alteration in specific microbiome clades, such as a HFD-dependent reduction in abundance of Bifidobacterium only in male mice. In knee synovium and infrapatellar fat tissue, HFD upregulated the expression of pro-inflammatory and pro-fibrotic genes predominantly in female mice. In cartilage, lipid metabolism proteins were more abundant with HFD in male mice, whereas proteins involved in glycolysis/gluconeogenesis and biosynthesis of amino acids were greater in cartilage of female mice. Sex-dependent metabolic differences were observed in cartilage from young, healthy mice prior to pubertal maturation, but not in primary juvenile chondrocytes studied in vitro. CONCLUSIONS HFD induced numerous sex differences in metabolic and inflammatory outcomes, especially in joint tissues, suggesting that sex-specific cellular processes are involved during development of early-stage OA with obesity.
Collapse
Affiliation(s)
- Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
- Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA.
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Erika Barboza Prado Lopes
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Labcorp Drug Development, Indianapolis, IN, USA
| | - Dominic Cortassa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- VA Oklahoma City Health Care, Oklahoma City, OK, USA
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Be'er Sheva, 84105, Israel
| | - Matlock A Jeffries
- Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Dawid Makosa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- University of Western Australia, Perth, Western Australia, Australia
| | - Anita Jopkiewicz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Panier Group, Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - Padmaja Mehta-D'souza
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Ravi K Komaravolu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Michael T Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| |
Collapse
|
15
|
Vishwanath K, McClure SR, Bonassar LJ. Heterogeneous distribution of viscosupplements in vivo is correlated to ex vivo frictional properties of equine cartilage. J Biomed Mater Res A 2024; 112:2149-2159. [PMID: 38923105 DOI: 10.1002/jbm.a.37766] [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: 03/02/2024] [Revised: 06/03/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
Intra-articular injections of hyaluronic acid (HA) are the cornerstone of osteoarthritis (OA) treatments. However, the mechanism of action and efficacy of HA viscosupplementation are debated. As such, there has been recent interest in developing synthetic viscosupplements. Recently, a synthetic 4 wt% polyacrylamide (pAAm) hydrogel was shown to effectively lubricate and bind to the surface of cartilage in vitro. However, its ability to localize to cartilage and alter the tribological properties of the tissue in a live articulating large animal joint is not known. The goal of this study was to quantify the distribution and extent of localization of pAAm in the equine metacarpophalangeal or metatarsophalangeal joint (fetlock joint), and determine whether preferential localization of pAAm influences the tribological properties of the tissue. An established planar fluorescence imaging technique was used to visualize and quantify the distribution of fluorescently labeled pAAm within the joint. While the pAAm hydrogel was present on all surfaces, it was not uniformly distributed, with more material present near the site of the injection. The lubricating ability of the cartilage in the joint was then assessed using a custom tribometer across two orders of magnitude of sliding speed in healthy synovial fluid. Cartilage regions with a greater coverage of pAAm, that is, higher fluorescent intensities, exhibited friction coefficients nearly 2-fold lower than regions with lesser pAAm (Rrm = -0.59, p < 0.001). Collectively, the findings from this study indicate that intra-articular viscosupplement injections are not evenly distributed inside a joint, and the tribological outcomes of these materials is strongly determined by the ability of the material to localize to the articulating surfaces in the joint.
Collapse
Affiliation(s)
- Karan Vishwanath
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York, USA
| | | | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
16
|
Scanzello CR, Hasty KA, Chung CB, Griffin TM, Willet NJ, Krug H, Chu CQ, Ewart D, Jerban S, Baker JF, Duvall CL, Brunger JM, Burdick JA, Spindler KP, Drissi H. Teaming up to overcome challenges toward translation of new therapeutics for osteoarthritis. J Orthop Res 2024; 42:2659-2672. [PMID: 39103981 PMCID: PMC12063556 DOI: 10.1002/jor.25944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024]
Abstract
As a leading global cause of musculoskeletal-related disability, osteoarthritis (OA) represents a public health urgency. Understanding of disease pathogenesis has advanced substantially in the past decade, yet no disease-modifying therapeutics have advanced to the clinic. To address this challenge, the CARE-AP (Cartilage Repair strategies to alleviate Arthritis Pain) collaborative research team was convened to bring together relevant multidisciplinary expertise and perspectives from across the VA research community nationwide. The first CARE-AP Annual Research Symposium took place (virtually) in February 2022 with roughly 90 participants. A number of innovative and therapeutic strategies were discussed, including siRNA approaches coupled with novel nanoparticle-based delivery systems, cellular engineering approaches to develop reparative cells that can probe the joint environment and respond to disease-specific cues, and novel biofabrication techniques to improve tissue engineering and effect "biological joint replacement." In addition, challenges and advances in rehabilitation approaches, imaging outcomes, and clinical studies were presented, which were integrated into a framework of recommendations for running "preclinical trials" to improve successful clinical translation.
Collapse
Affiliation(s)
- Carla R. Scanzello
- Translational Musculoskeletal Research Center, Corp. Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Karen A. Hasty
- Research Service 151, Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, Tennessee, USA
- Department of Orthopaedic Surgery and Biomedical Engineering, Campbell Clinic/University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Christine B. Chung
- Radiology Service, Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Timothy M. Griffin
- Oklahoma City VA Health Care System, Oklahoma City, Oklahoma, USA
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Nick J. Willet
- Veterans Affairs Portland Health Care System, Portland, Oregon, USA
- Phil and Penny Knight Campus for Accelerating Scientific Impact, Department of Bioengineering, University of Oregon, Eugene, Oregon, USA
| | - Hollis Krug
- Rheumatology Section, Minneapolis Veterans Affairs Medical Center, Minneapolis, Minnesota, USA
- Division of Rheumatology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cong-Qiu Chu
- Veterans Affairs Portland Health Care System, Portland, Oregon, USA
- Division of Arthritis and Rheumatic Diseases, Oregon Health Sciences University, Portland, Oregon, USA
| | - David Ewart
- Rheumatology Section, Minneapolis Veterans Affairs Medical Center, Minneapolis, Minnesota, USA
- Division of Rheumatology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Saeed Jerban
- Radiology Service, Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Joshua F. Baker
- Translational Musculoskeletal Research Center, Corp. Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jonathan M. Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Jason A. Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, USA
| | - Kurt P. Spindler
- Department of Orthopaedic Surgery, Sports Medicine, Cleveland Clinic Florida, Coral Springs, Florida, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta VA Medical Center, Decatur, Georgia, USA
| |
Collapse
|
17
|
Oláh T, Cucchiarini M, Madry H. Temporal progression of subchondral bone alterations in OA models involving induction of compromised meniscus integrity in mice and rats: A scoping review. Osteoarthritis Cartilage 2024; 32:1220-1234. [PMID: 38876436 DOI: 10.1016/j.joca.2024.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024]
Abstract
OBJECTIVE To categorize the temporal progression of subchondral bone alterations induced by compromising meniscus integrity in mouse and rat models of knee osteoarthritis (OA). METHOD Scoping review of investigations reporting subchondral bone changes with appropriate negative controls in the different mouse and rat models of OA induced by compromising meniscus integrity. RESULTS The available literature provides appropriate temporal detail on subchondral changes in these models, covering the entire spectrum of OA with an emphasis on early and mid-term time points. Microstructural changes of the subarticular spongiosa are comprehensively described; those of the subchondral bone plate are not. In mouse models, global subchondral bone alterations are unidirectional, involving an advancing sclerosis of the trabecular structure over time. In rats, biphasic subchondral bone alterations begin with an osteopenic degeneration and loss of subchondral trabeculae, progressing to a late sclerosis of the entire subchondral bone. Rat models, independently from the applied technique, relatively faithfully mirror the early bone loss detected in larger animals, and the late subchondral bone sclerosis observed in human advanced OA. CONCLUSION Mice and rats allow us to study the microstructural consequences of compromising meniscus integrity at high temporal detail. Thickening of the subchondral bone plate, an early loss of thinner subarticular trabecular elements, followed by a subsequent sclerosis of the entire subchondral bone are all important and reliable hallmarks that occur in parallel with the advancing articular cartilage degeneration. Thoughtful decisions on the study design, laterality, selection of controls and volumes of interest are crucial to obtain meaningful data.
Collapse
Affiliation(s)
- Tamás Oláh
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany; Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany.
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany.
| |
Collapse
|
18
|
Vishwanath K, Secor EJ, Watkins A, Reesink HL, Bonassar LJ. Loss of effective lubricating viscosity is the primary mechanical marker of joint inflammation in equine synovitis. J Orthop Res 2024; 42:1438-1447. [PMID: 38291343 DOI: 10.1002/jor.25793] [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: 06/27/2023] [Revised: 11/17/2023] [Accepted: 12/24/2023] [Indexed: 02/01/2024]
Abstract
Inflammation of the synovium, known as synovitis, plays an important role in the pathogenesis of osteoarthritis (OA). Synovitis involves the release of a wide variety of pro-inflammatory mediators in synovial fluid (SF) that damage the articular cartilage extracellular matrix and induce death and apoptosis in chondrocytes. The composition of synovial fluid is dramatically altered by inflammation in OA, with changes to both hyaluronic acid and lubricin, the primary lubricating molecules in SF. However, the relationship between key biochemical markers of joint inflammation and mechanical function of SF is not well understood. Here, we demonstrate the application of a novel analytical framework to measure the effective viscosity for SF lubrication of cartilage, which is distinct from conventional rheological viscosity. Notably, in a well-established equine model of synovitis, this effective lubricating viscosity decreased by up to 10,000-fold for synovitis SF compared to a ~4 fold change in conventional viscosity measurements. Further, the effective lubricating viscosity was strongly inversely correlated (r = -0.6 to -0.8) to multiple established biochemical markers of SF inflammation, including white blood cell count, prostaglandin E2 (PGE2), and chemokine ligand (CCLs) concentrations, while conventional measurements of viscosity were poorly correlated to these markers. These findings demonstrate the importance of experimental and analytical approaches to characterize functional lubricating properties of synovial fluid and their relationships to soluble biomarkers to better understand the progression of OA.
Collapse
Affiliation(s)
- Karan Vishwanath
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York, USA
| | - Erica J Secor
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Amanda Watkins
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Heidi L Reesink
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
19
|
Ma Z, Li DX, Lan X, Bubelenyi A, Vyhlidal M, Kunze M, Sommerfeldt M, Adesida AB. Short-term response of primary human meniscus cells to simulated microgravity. Cell Commun Signal 2024; 22:342. [PMID: 38907358 PMCID: PMC11191296 DOI: 10.1186/s12964-024-01684-w] [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: 02/22/2024] [Accepted: 05/27/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Mechanical unloading of the knee articular cartilage results in cartilage matrix atrophy, signifying the osteoarthritic-inductive potential of mechanical unloading. In contrast, mechanical loading stimulates cartilage matrix production. However, little is known about the response of meniscal fibrocartilage, a major mechanical load-bearing tissue of the knee joint, and its functional matrix-forming fibrochondrocytes to mechanical unloading events. METHODS In this study, primary meniscus fibrochondrocytes isolated from the inner avascular region of human menisci from both male and female donors were seeded into porous collagen scaffolds to generate 3D meniscus models. These models were subjected to both normal gravity and mechanical unloading via simulated microgravity (SMG) for 7 days, with samples collected at various time points during the culture. RESULTS RNA sequencing unveiled significant transcriptome changes during the 7-day SMG culture, including the notable upregulation of key osteoarthritis markers such as COL10A1, MMP13, and SPP1, along with pathways related to inflammation and calcification. Crucially, sex-specific variations in transcriptional responses were observed. Meniscus models derived from female donors exhibited heightened cell proliferation activities, with the JUN protein involved in several potentially osteoarthritis-related signaling pathways. In contrast, meniscus models from male donors primarily regulated extracellular matrix components and matrix remodeling enzymes. CONCLUSION These findings advance our understanding of sex disparities in knee osteoarthritis by developing a novel in vitro model using cell-seeded meniscus constructs and simulated microgravity, revealing significant sex-specific molecular mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Zhiyao Ma
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - David Xinzheyang Li
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Department of Civil and Environmental Engineering, Faculty of Engineering, AB, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Xiaoyi Lan
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Adam Bubelenyi
- Faculty of Science, AB, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Margaret Vyhlidal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Melanie Kunze
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Mark Sommerfeldt
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Adetola B Adesida
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada.
- Department of Mechanical Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, T6G 2R3, Canada.
| |
Collapse
|
20
|
Chapman JH, Ghosh D, Attari S, Ude CC, Laurencin CT. Animal Models of Osteoarthritis: Updated Models and Outcome Measures 2016-2023. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:127-146. [PMID: 38983776 PMCID: PMC11233113 DOI: 10.1007/s40883-023-00309-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2024]
Abstract
Purpose Osteoarthritis (OA) is a global musculoskeletal disorder that affects primarily the knee and hip joints without any FDA-approved disease-modifying therapies. Animal models are essential research tools in developing therapies for OA; many animal studies have provided data for the initiation of human clinical trials. Despite this, there is still a need for strategies to recapitulate the human experience using animal models to better develop treatments and understand pathogenesis. Since our last review on animal models of osteoarthritis in 2016, there have been exciting updates in OA research and models. The main purpose of this review is to update the latest animal models and key features of studies in OA research. Method We used our existing classification method and screened articles in PubMed and bibliographic search for animal OA models between 2016 and 2023. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Recent studies were analyzed and classified. We also identified ex vivo models as an area of ongoing research. Each animal model offers its own benefit in the study of OA and there are a full range of outcome measures that can be assessed. Despite the vast number of models, each has its drawbacks that have limited translating approved therapies for human use. Conclusion Depending on the outcome measures and objective of the study, researchers should pick the best model for their work. There have been several exciting studies since 2016 that have taken advantage of regenerative engineering techniques to develop therapies and better understand OA. Lay Summary Osteoarthritis (OA) is a chronic debilitating disease without any cure that affects mostly the knee and hip joints and often results in surgical joint replacement. Cartilage protects the joint from mechanical forces and degrades with age or in response to injury. The many contributing causes of OA are still being investigated, and animals are used for preclinical research and to test potential new treatments. A single consensus OA animal model for preclinical studies is non-existent. In this article, we review the many animal models for OA and provide a much-needed update on studies and model development since 2016.
Collapse
Affiliation(s)
- James H. Chapman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Seyyedmorteza Attari
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Chinedu C. Ude
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical and Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
21
|
Stewart HL, Gilbert D, Stefanovski D, Garman Z, Albro MB, Bais M, Grinstaff MW, Snyder BD, Schaer TP. A missed opportunity: A scoping review of the effect of sex and age on osteoarthritis using large animal models. Osteoarthritis Cartilage 2024; 32:501-513. [PMID: 38408635 PMCID: PMC11534084 DOI: 10.1016/j.joca.2024.02.009] [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: 10/27/2023] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
OBJECTIVE The objective was to critically analyze the published literature accounting for sex differences and skeletal age (open vs. closed physis) in preclinical animal models of OA, including the disaggregation of data by sex and skeletal maturity when data is generated from combined sex and/or multi-aged cohorts without proper confounding. METHOD A scoping literature review of PubMed, Web of Science, EMBASE, and SCOPUS was performed for studies evaluating the effect of sex and age in experimental studies and clinical trials utilizing preclinical large animal models of OA. RESULTS A total of 9727 papers were identified in large animal (dog, pig, sheep, goat, horse) models for preclinical OA research, of which 238 ex vivo and/or in vivo studies disclosed model type, animal species, sex, and skeletal age sufficient to analyze their effect on outcomes. Dogs, followed by pigs, sheep, and horses, were the most commonly used models. A paucity of preclinical studies evaluated the effect of sex and age in large animal models of naturally occurring or experimentally induced OA: 26 total studies reported some kind of analysis of the effects of sex or age, with 4 studies discussing the effects of sex only, 11 studies discussing the effects of age only, and 11 studies analyzing both the effects of age and sex. CONCLUSION Fundamental to translational research, OARSI is uniquely positioned to develop recommendations for conducting preclinical studies using large animal models of OA that consider biological mechanisms linked to sex chromosomes, skeletal age, castration, and gonadal hormones affecting OA pathophysiology and treatment response.
Collapse
Affiliation(s)
- Holly L Stewart
- Department of Clinical Studies New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, PA 19348, USA
| | - Derek Gilbert
- Department of Clinical Studies New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, PA 19348, USA
| | - Darko Stefanovski
- Department of Clinical Studies New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, PA 19348, USA
| | - Zoe Garman
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston MA 02215, USA
| | - Michael B Albro
- Department of Mechanical Engineering, Boston University, Boston MA 02215, USA
| | - Manish Bais
- Boston University, Henry M. Goldman School of Dental Medicine, Boston MA 02118, USA
| | - Mark W Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston MA 02215, USA
| | - Brian D Snyder
- Department of Orthopaedic Surgery, Boston Children's Hospital Boston, MA 02215, USA
| | - Thomas P Schaer
- Department of Clinical Studies New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, PA 19348, USA.
| |
Collapse
|
22
|
Adam AO, Benea HRC, Fotescu HM, Alcalá Ruiz M, Cimpean GC, Ciornei V, Cernacovschi A, Edves AR, Crisan M. Recent Trends in Adipose Tissue-Derived Injectable Therapies for Osteoarthritis: A Scoping Review of Animal Models. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:707. [PMID: 38792890 PMCID: PMC11123108 DOI: 10.3390/medicina60050707] [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: 02/01/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024]
Abstract
Background and Objectives: This scoping review investigates recent trends in adipose tissue-derived injectable therapies for osteoarthritis (OA) in animal models, focusing on minimally manipulated or lightly processed adipose tissue. By evaluating and examining the specific context in which these therapies were investigated across diverse animal OA models, this review aims to provide valuable insights that will inform and guide future research and clinical applications in the ongoing pursuit of effective treatments for osteoarthritis. Materials and Methods: This research conducted a comprehensive literature review of PubMed and Embase to determine studies about minimally manipulated adipose tissue-derived injectable therapies for osteoarthritis investigated using animal models. The primary search found 530 results. After excluding articles that focused on spontaneous osteoarthritis; on transfected, preconditioned, cultured, or co-cultured adipose-derived stem cells; and articles with unavailable full text, we included 11 articles in our review. Results: The examined therapies encompassed mechanical micro-fragmented adipose tissue (MFAT) and stromal vascular fraction (SVF) obtained via collagenase digestion and centrifugation. These interventions were evaluated across various animal models, including mice, rats, rabbits, and sheep with induced OA. Notably, more studies concentrated on surgically induced OA rather than chemically induced OA. The assessment of these therapies focused on elucidating their protective immunomodulatory, anti-inflammatory, and chondroregenerative potential through comprehensive evaluations, including macroscopic assessments, histological analyses, immunohistochemical examinations, and biochemical assays. Conclusions: This review provides a comprehensive analysis of adipose tissue-derived injectable therapies for osteoarthritis across diverse animal models. While revealing potential benefits and insights, the heterogeneity of data and the limited number of studies highlight the need for further research to formulate conclusive recommendations for clinical applications.
Collapse
Affiliation(s)
- Alina Otilia Adam
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Horea Rares Ciprian Benea
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Horia Mihnea Fotescu
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Miriam Alcalá Ruiz
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - George Claudiu Cimpean
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Vladimir Ciornei
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Arsenii Cernacovschi
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Andrei Rares Edves
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Maria Crisan
- Department of Histology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| |
Collapse
|
23
|
Bei M, Zheng Z, Xiao Y, Liu N, Cao X, Tian F, Zhang L, Wu X. Effects of alendronate on cartilage lesions and micro-architecture deterioration of subchondral bone in patellofemoral osteoarthritic ovariectomized rats with patella-baja. J Orthop Surg Res 2024; 19:197. [PMID: 38528611 DOI: 10.1186/s13018-024-04677-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/14/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Patellofemoral osteoarthritis (PFJOA) is a subtype of knee OA, which is one of the main causes of anterior knee pain. The current study found an increased prevalence of OA in postmenopausal women, called postmenopausal OA. Therefore, we designed the ovariectomized rat model of patella baja-induced PFJOA. Alendronate (ALN) inhibits osteoclast-mediated bone loss, and has been reported the favorable result of a potential intervention option of OA treatment. However, the potential effects of ALN treatment on PFJOA in the ovariectomized rat model are unknown and need further investigation prior to exploration in the clinical research setting. In this study, the effects of ALN on articular cartilage degradation and subchondral bone microstructure were assessed in the ovariectomized PFJOA rat model for 10 weeks. METHODS Patella baja and estrogen withdrawal were induced by patellar ligament shortening (PLS) and bilateral ovariectmomy surgeries in 3-month-old female Sprague-Dawley rats, respectively. Rats were randomly divided into five groups (n = 8): Sham + V; OVX + V, Sham + PLS + V, OVX + PLS + V, OVX + PLS + ALN (ALN: 70 μg/kg/week). Radiography was performed to evaluate patellar height ratios, and the progression of PFJOA was assessed by macroscopic and microscopic analyses, immunohistochemistry and micro-computed tomography (micro-CT). RESULTS Our results found that the patella baja model prepared by PLS can successfully cause degeneration of articular cartilage and subchondral bone, resulting in changes of PFJOA. OVX caused a decrease in estrogen levels in rats, which aggravated the joint degeneration caused by PFJOA. Early application of ALN can delay the degenerative changes of articular cartilage and subchondral bone microstructure in castrated PFJOA rat to a certain extent, improve and maintain the micrometabolism and structural changes of cartilage and subchondral bone. CONCLUSION The early application of ALN can delay the destruction of articular cartilage and subchondral bone microstructure in castrated PFJOA rat to a certain extent.
Collapse
Affiliation(s)
- Mingjian Bei
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Xinjiekoudongjie 31, Xicheng Dis, Beijing, 100035, People's Republic of China
| | - Zhiyuan Zheng
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, People's Republic of China
| | - Yaping Xiao
- The Department of Orthopedic Surgery, Wuhan Third Hospital, Tongren Hospital of Wuhan University, No. 241, Pengliuyang Road, Wuhan, 430000, People's Republic of China
| | - Ning Liu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, People's Republic of China
| | - Xuehui Cao
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, People's Republic of China
| | - Faming Tian
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, People's Republic of China
| | - Liu Zhang
- Department of Orthopedic Surgery, Emergency General Hospital, Xibahenanli 29, Chaoyang District, Beijing, 100028, People's Republic of China
| | - Xinbao Wu
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Xinjiekoudongjie 31, Xicheng Dis, Beijing, 100035, People's Republic of China.
| |
Collapse
|
24
|
Jiang S, Xie W, Knapstein PR, Donat A, Albertsen LC, Sevecke J, Erdmann C, Appelt J, Fuchs M, Hildebrandt A, Maleitzke T, Frosch KH, Baranowsky A, Keller J. Transcript-dependent effects of the CALCA gene on the progression of post-traumatic osteoarthritis in mice. Commun Biol 2024; 7:223. [PMID: 38396204 PMCID: PMC10891124 DOI: 10.1038/s42003-024-05889-0] [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: 05/26/2023] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Osteoarthritis represents a chronic degenerative joint disease with exceptional clinical relevance. Polymorphisms of the CALCA gene, giving rise to either a procalcitonin/calcitonin (PCT/CT) or a calcitonin gene-related peptide alpha (αCGRP) transcript by alternative splicing, were reported to be associated with the development of osteoarthritis. The objective of this study was to investigate the role of both PCT/CT and αCGRP transcripts in a mouse model of post-traumatic osteoarthritis (ptOA). WT, αCGRP-/- and CALCA-/- mice were subjected to anterior cruciate ligament transection (ACLT) to induce ptOA of the knee. Mice were sacrificed 4 and 8 weeks post-surgery, followed by micro-CT and histological evaluation. Here we show that the expression of both PCT/CT and αCGRP transcripts is induced in ptOA knees. CALCA-/- mice show increased cartilage degeneration and subchondral bone loss with elevated osteoclast numbers compared to αCGRP-/- and WT mice. Osteophyte formation is reduced to the same extent in CALCA-/- and αCGRP-/- mice compared to WT controls, while a reduced synovitis score is noticed exclusively in mice lacking CALCA. Our data show that expression of the PCT/CT transcript protects from the progression of ptOA, while αCGRP promotes osteophyte formation, suggesting that CALCA-encoded peptides may represent novel targets for the treatment of ptOA.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Weixin Xie
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Richard Knapstein
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Donat
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lilly-Charlotte Albertsen
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Sevecke
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cordula Erdmann
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jessika Appelt
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Melanie Fuchs
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Alexander Hildebrandt
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
| | - Tazio Maleitzke
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
- Department of Orthopaedic Surgery, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Karl-Heinz Frosch
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Trauma Surgery, Orthopedics and Sports Traumatology, BG Hospital Hamburg, Hamburg, Germany
| | - Anke Baranowsky
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
25
|
Fernández-Moreno M, Hermida-Gómez T, Larkins N, Reynolds A, Blanco FJ. Anti-Inflammatory Activity of APPA (Apocynin and Paeonol) in Human Articular Chondrocytes. Pharmaceuticals (Basel) 2024; 17:118. [PMID: 38256951 PMCID: PMC10818286 DOI: 10.3390/ph17010118] [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: 12/13/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease leading to cartilage loss and reduction in the joint space which results in pain. The current pharmacological treatment of OA is inadequate and pharmacological interventions focus on symptom management. APPA, a combination of apocynin (AP) and paeonol (PA), is a potential drug for treating OA. The aim of this study was to analyze the effects of APPA on the modulation of the inflammatory response in chondrocytes. Samples were incubated with IL-1β and APPA, and their responses to proinflammatory cytokines, catabolic mediators and redox responses were then measured. The effect of APPA on mitogenesis was also evaluated. Results show that APPA attenuated the expression of IL-8, TNF-α, MMP-3, MMP-13, SOD-2 and iNOS, resulting in the protection of human articular cartilage. APPA decreased PGC-1α gene expression induced by IL-1β. APPA did not modulate the gene expression of Mfn2, Sirt-1 or Sirt-3. The overall findings indicate that APPA may be an effective treatment for OA by targeting several of the pathways involved in OA pathogenesis.
Collapse
Affiliation(s)
- Mercedes Fernández-Moreno
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade de A Coruña (UDC), 15071 A Coruña, Spain;
- Grupo de Investigación en Reumatología y Salud (GIR-S), Centro Interdisciplinar de Química y Biología (CICA), Universidade de A Coruña (UDC), Campus de Elviña, 15071 A Coruña, Spain
| | - Tamara Hermida-Gómez
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade de A Coruña (UDC), 15071 A Coruña, Spain;
- Grupo de Investigación en Reumatología y Salud (GIR-S), Centro Interdisciplinar de Química y Biología (CICA), Universidade de A Coruña (UDC), Campus de Elviña, 15071 A Coruña, Spain
- Centro de Investigación Biomédica en Red, Bioingenieria, Biomatereial y Nanomedicina (CIBER-BBN), 50018 Zaragoza, Spain
| | - Nicholas Larkins
- AKL Therapeutics Ltd., Stevenage Bioscience, Gunnels Wood Rd, Stevenage SG1 2FX, UK; (N.L.); (A.R.)
| | - Alan Reynolds
- AKL Therapeutics Ltd., Stevenage Bioscience, Gunnels Wood Rd, Stevenage SG1 2FX, UK; (N.L.); (A.R.)
| | - Francisco J. Blanco
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade de A Coruña (UDC), 15071 A Coruña, Spain;
- Grupo de Investigación en Reumatología y Salud (GIR-S), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Centro Interdisciplinar de Química y Biología (CICA), INIBIC-Sergas, Universidade de A Coruña (UDC), Campus de Oza, 15008 A Coruña, Spain
| |
Collapse
|
26
|
Timkovich AE, Holling GA, Afzali MF, Kisiday J, Santangelo KS. TLR4 antagonism provides short-term but not long-term clinical benefit in a full-depth cartilage defect mouse model. Connect Tissue Res 2024; 65:26-40. [PMID: 37898909 PMCID: PMC11271750 DOI: 10.1080/03008207.2023.2269257] [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: 12/14/2022] [Revised: 08/23/2023] [Accepted: 10/05/2023] [Indexed: 10/31/2023]
Abstract
PURPOSE/AIM Cartilage injury and subsequent osteoarthritis (OA) are debilitating conditions affecting millions worldwide. As there are no cures for these ailments, novel therapies are needed to suppress disease pathogenesis. Given that joint injuries are known to produce damage-associated molecular patterns (DAMPs), our central premise is that the Toll-like receptor 4 (TLR4) pathway is a principal driver in the early response to cartilage damage and subsequent pathology. We postulate that TLR4 activation is initiated/perpetuated by DAMPs released following joint damage. Thus, antagonism of the TLR4 pathway immediately after injury may suppress the development of joint surface defects. MATERIALS AND METHODS Two groups were utilized: (1) 8-week-old, male C57BL6 mice treated systemically with a known TLR4 antagonist and (2) mice injected with vehicle control. A full-depth cartilage lesion on the midline of the patellofemoral groove was created in the right knee of each mouse. The left knee was used as a sham surgery control. Gait changes were evaluated over 4 weeks using a quantitative gait analysis system. At harvest, knee joints were processed for pathologic assessment, Nanostring® transcript expression, and immunohistochemistry (IHC). RESULTS Short-term treatment with a TLR4 antagonist at 14-days significantly improved relevant gait parameters; improved cartilage metrics and modified Mankin scores were also seen. Additionally, mRNA expression and IHC showed reduced expression of inflammatory mediators in animals treated with the TLR4 antagonist. CONCLUSIONS Collectively, this work demonstrates that systemic treatment with a TLR4 antagonist is protective to further cartilage damage 14-days post-injury in a murine model of induced disease.
Collapse
Affiliation(s)
- Ariel E. Timkovich
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - G. Aaron Holling
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Maryam F. Afzali
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - John Kisiday
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Kelly S. Santangelo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
27
|
Haubruck P, Heller R, Blaker CL, Clarke EC, Smith SM, Burkhardt D, Liu Y, Stoner S, Zaki S, Shu CC, Little CB. Streamlining quantitative joint-wide medial femoro-tibial histopathological scoring of mouse post-traumatic knee osteoarthritis models. Osteoarthritis Cartilage 2023; 31:1602-1611. [PMID: 37716405 DOI: 10.1016/j.joca.2023.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 07/03/2023] [Accepted: 07/26/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVES Histological scoring remains the gold-standard for quantifying post-traumatic osteoarthritis (ptOA) in animal models, allowing concurrent evaluation of numerous joint tissues. Available systems require scoring multiple sections/joint making analysis laborious and expensive. We investigated if a single section allowed equivalent quantitation of pathology in different joint tissues and disease stages, in three ptOA models. METHOD Male 10-12-week-old C57BL/6 mice underwent surgical medial-meniscal-destabilization, anterior-cruciate-ligament (ACL) transection, non-invasive-ACL-rupture, or served as sham-surgical, non-invasive-ACL-strain, or naïve/non-operated controls. Mice (n = 12/group) were harvested 1-, 4-, 8-, and 16-week post-intervention. Serial sagittal toluidine-blue/fast-green stained sections of the medial-femoro-tibial joint (n = 7/joint, 84 µm apart) underwent blinded scoring of 40 histology-outcomes. We evaluated agreement between single-slide versus entire slide-set maximum or median scores (weighted-kappa), and sensitivity/specificity of single-slide versus median/maximum to detect OA pathology. RESULTS A single optimal mid-sagittal section showed excellent agreement with median (weighted-kappa 0.960) and maximum (weighted-kappa 0.926) scores. Agreement for individual histology-outcomes was high with only 19/240 median and 15/240 maximum scores having a weighted-kappa ≤0.4, the majority of these (16/19 and 11/15) in control groups. Statistically-significant histology-outcome differences between ptOA models and their controls detected with the entire slide-set were reliably reproduced using a single slide (sensitivity >93.15%, specificity >93.10%). The majority of false-negatives with single-slide scoring were meniscal and subchondral bone histology-outcomes (89%) and occurred in weeks 1-4 post-injury (84%). CONCLUSION A single mid-sagittal slide reduced the time needed to score diverse histopathological changes by 87% without compromising the sensitivity or specificity of the analysis, across a variety of ptOA models and time-points.
Collapse
Affiliation(s)
- Patrick Haubruck
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, D-69118 Heidelberg, Germany; Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Raban Heller
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, D-69118 Heidelberg, Germany; Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany; Bundeswehr Hospital Berlin, Clinic of Traumatology and Orthopaedics, D-10115 Berlin, Germany
| | - Carina L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia; Murray Maxwell Biomechanics Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Elizabeth C Clarke
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Susan M Smith
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Daniel Burkhardt
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Yolanda Liu
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Shihani Stoner
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Sanaa Zaki
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia; Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia
| | - Cindy C Shu
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| |
Collapse
|
28
|
Macfarlane E, Cavanagh L, Fong-Yee C, Tuckermann J, Chen D, Little CB, Seibel MJ, Zhou H. Deletion of the chondrocyte glucocorticoid receptor attenuates cartilage degradation through suppression of early synovial activation in murine posttraumatic osteoarthritis. Osteoarthritis Cartilage 2023; 31:1189-1201. [PMID: 37105394 DOI: 10.1016/j.joca.2023.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/31/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVE Disruption of endogenous glucocorticoid signalling in bone cells attenuates osteoarthritis (OA) in aged mice, however, the role of endogenous glucocorticoids in chondrocytes is unknown. Here, we investigated whether deletion of the glucocorticoid receptor, specifically in chondrocytes, also alters OA progression. DESIGN Knee OA was induced by surgical destabilisation of the medial meniscus (DMM) in male 22-week-old tamoxifen-inducible glucocorticoid receptor knockout (chGRKO) mice and their wild-type (WT) littermates (n = 7-9/group). Mice were harvested 2, 4, 8 and 16 weeks after surgery to examine the spatiotemporal changes in molecular, cellular, and histological characteristics. RESULTS At all time points following DMM, cartilage damage was significantly attenuated in chGRKO compared to WT mice. Two weeks after DMM, WT mice exhibited increased chondrocyte and synoviocyte hypoxia inducible factor (HIF)-2α expression resulting in extensive synovial activation characterised by synovial thickening and increased interleukin-1 beta expression. At 2 and 4 weeks after DMM, WT mice displayed pronounced chondrocyte senescence and elevated catabolic signalling (reduced Yes-associated protein 1 (YAP1) and increased matrix metalloprotease [MMP]-13 expression). Contrastingly, at 2 weeks after DMM, HIF-2α expression and synovial activation were much less pronounced in chGRKO than in WT mice. Furthermore, chondrocyte YAP1 and MMP-13 expression, as well as chondrocyte senescence were similar in chGRKO-DMM mice and sham-operated controls. CONCLUSION Endogenous glucocorticoid signalling in chondrocytes promotes synovial activation, chondrocyte senescence and cartilage degradation by upregulation of catabolic signalling through HIF-2α in murine posttraumatic OA. These findings indicate that inhibition of glucocorticoid signalling early after injury may present a promising way to slow osteoarthritic cartilage degeneration.
Collapse
Affiliation(s)
- Eugenie Macfarlane
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia.
| | - Lauryn Cavanagh
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia.
| | - Colette Fong-Yee
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia.
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Baden-Württemberg, Germany.
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| | - Christopher B Little
- Raymond Purves Laboratories, Kolling Institute and Institute of Bone and Joint Research, University of Sydney, and Royal North Shore Hospital, St. Leonards, NSW, Australia.
| | - Markus J Seibel
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia; Department of Endocrinology and Metabolism, Concord Repatriation General Hospital, Sydney, NSW, Australia.
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
29
|
Hong Y, Johnston L, Wang W. A Multi-factor Analysis of Revision in Total Hip Replacement Using the Collarless-Polished-Tapered Stems with Different Cups. Orthop Surg 2023. [PMID: 37385946 DOI: 10.1111/os.13778] [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: 02/14/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 07/01/2023] Open
Abstract
OBJECTIVE Collarless-polished-tapered (CPT) stems have been widely used in total hip replacement (THR). Given that various types of cups are combined with CPT in clinical practice, however, what cup type performs the best for use with CPT is still unclear. This study aimed to investigate the effects of three types of commonly used cups with CPT on revision and survival life using multi-factor analysis. METHODS This study is a cohort study using the data between October 1998 to September 2021. The data of THR patients with ZCA All-poly Acetabular Cup, Continuum Acetabular System, and Trilogy Acetabular System with CPT were collected from several hospitals in the UK. The patients aged from 20 to 97 (n = 5981, 2345 male and 3636 female). Age, gender, body mass index, diagnosis, surgeon grade, cup material, cup size, surgical approach, survival life, complications, and Harris hip scores (HHS) were analyzed in relation to revision status. SPSS statistical software was used to analyze the relationship among various factors. The main statistical methods included chi-square with cross tables, analysis of variance (ANOVA) and survival analysis. RESULTS The results in relation to HHS shows that the continuum cup has the best outcome in the postoperative period of 1 and 5 years (1 year = 90.7, 5 years = 91.3; P < 0.001); the Trilogy cup was the second (1 year = 88.4, 5 years = 87.3; P < 0.001); and the ZCA cup was the third (1 year = 84.6, 5 years = 82.4; P < 0.001). However, the Trilogy cup performed the best regarding survival life on revision while the Continuum cup was the worst. CONCLUSION When the CPT stem is combined with different cups, the trilogy cup shows the best characteristics in terms of survival trends with revision ratios compared with the continuum and ZCA cups, and is therefore recommended by this study.
Collapse
Affiliation(s)
- Yu Hong
- University Department of Orthopaedic and Trauma Surgery, Ninewells Hospital and Medical School, TORT Centre, University of Dundee, Dundee, UK
| | - Linda Johnston
- University Department of Orthopaedic and Trauma Surgery, Ninewells Hospital and Medical School, TORT Centre, University of Dundee, Dundee, UK
| | - Weijie Wang
- University Department of Orthopaedic and Trauma Surgery, Ninewells Hospital and Medical School, TORT Centre, University of Dundee, Dundee, UK
| |
Collapse
|
30
|
Tang Y, Hong F, Ding S, Yang J, Zhang M, Ma Y, Zheng Q, Yang D, Jin Y, Ma C. METTL3-mediated m 6A modification of IGFBP7-OT promotes osteoarthritis progression by regulating the DNMT1/DNMT3a-IGFBP7 axis. Cell Rep 2023; 42:112589. [PMID: 37270777 DOI: 10.1016/j.celrep.2023.112589] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/15/2023] [Accepted: 05/16/2023] [Indexed: 06/06/2023] Open
Abstract
Osteoarthritis (OA) is the most common degenerative disorder, affecting approximately half of the elderly population. In this study, we find that the expressions of long noncoding RNA (lncRNA) IGFBP7-OT and its maternal gene, IGFBP7, are upregulated and positively correlated in osteoarthritic cartilage. Overexpression of IGFBP7-OT significantly inhibits chondrocyte viability, promotes chondrocyte apoptosis, and reduces extracellular matrix components, whereas IGFBP7-OT knockdown has the opposite effects. IGFBP7-OT overexpression promotes cartilage degeneration and markedly aggravates the monosodium iodoacetate-induced OA phenotype in vivo. Further mechanistic research reveals that IGFBP7-OT promotes OA progression by upregulating IGFBP7 expression. Specifically, IGFBP7-OT suppresses the occupancy of DNMT1 and DNMT3a on the IGFBP7 promoter, thereby inhibiting methylation of the IGFBP7 promoter. The upregulation of IGFBP7-OT in OA is partially controlled by METTL3-mediated N6-methyladenosine (m6A) modification. Collectively, our findings reveal that m6A modification of IGFBP7-OT promotes OA progression by regulating the DNMT1/DNMT3a-IGFBP7 axis and provide a potential therapeutical target for OA treatment.
Collapse
Affiliation(s)
- Yuting Tang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China
| | - Fangling Hong
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China
| | - Siyang Ding
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China
| | - Jiashu Yang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China
| | - Ming Zhang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China
| | - Yunfei Ma
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China
| | - Que Zheng
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China
| | - Dawei Yang
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing, P.R. China
| | - Yucui Jin
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China.
| | - Changyan Ma
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing 211166, P.R. China.
| |
Collapse
|
31
|
Li Z, Lu J. CircRNAs in osteoarthritis: research status and prospect. Front Genet 2023; 14:1173812. [PMID: 37229197 PMCID: PMC10203419 DOI: 10.3389/fgene.2023.1173812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/18/2023] [Indexed: 05/27/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disease globally, and its progression is irreversible. The mechanism of osteoarthritis is not fully understood. Research on the molecular biological mechanism of OA is deepening, among which epigenetics, especially noncoding RNA, is an emerging hotspot. CircRNA is a unique circular noncoding RNA not degraded by RNase R, so it is a possible clinical target and biomarker. Many studies have found that circRNAs play an essential role in the progression of OA, including extracellular matrix metabolism, autophagy, apoptosis, the proliferation of chondrocytes, inflammation, oxidative stress, cartilage development, and chondrogenic differentiation. Differential expression of circRNAs was also observed in the synovium and subchondral bone in the OA joint. In terms of mechanism, existing studies have mainly found that circRNA adsorbs miRNA through the ceRNA mechanism, and a few studies have found that circRNA can serve as a scaffold for protein reactions. In terms of clinical transformation, circRNAs are considered promising biomarkers, but no large cohort has tested their diagnostic value. Meanwhile, some studies have used circRNAs loaded in extracellular vesicles for OA precision medicine. However, there are still many problems to be solved in the research, such as the role of circRNA in different OA stages or OA subtypes, the construction of animal models of circRNA knockout, and more research on the mechanism of circRNA. In general, circRNAs have a regulatory role in OA and have particular clinical potential, but further studies are needed in the future.
Collapse
Affiliation(s)
- Zhuang Li
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jun Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
32
|
Kim SJ, Kim JE, Choe G, Song DH, Kim SJ, Kim TH, Yoo J, Kim SH, Jung Y. Self-assembled peptide-substance P hydrogels alleviate inflammation and ameliorate the cartilage regeneration in knee osteoarthritis. Biomater Res 2023; 27:40. [PMID: 37143133 PMCID: PMC10161637 DOI: 10.1186/s40824-023-00387-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Self-assembled peptide (SAP)-substance P (SP) hydrogels can be retained in the joint cavity longer than SP alone, and they can alleviate inflammation and ameliorate cartilage regeneration in knee osteoarthritis (OA). We conducted a preclinical study using diverse animal models of OA and an in vitro study using human synoviocytes and patient-derived synovial fluids to demonstrate the effect of SAP-SP complex on the inflammation and cartilage regeneration. METHODS Surgical induction OA model was prepared with New Zealand white female rabbits and chemical induction, and naturally occurring OA models were prepared using Dunkin Hartely female guinea pigs. The SAP-SP complex or control (SAP, SP, or saline) was injected into the joint cavities in each model. We performed micro-computed tomography (Micro-CT) analysis, histological evaluation, immunofluorescent analysis, and terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling (TUNEL) assay and analyzed the recruitment of intrinsic mesenchymal stem cells (MSCs), macrophage activity, and inflammatory cytokine in each OA model. Human synoviocytes were cultured in synovial fluid extracted from human OA knee joints injected with SAP-SP complexes or other controls. Proliferative capacity and inflammatory cytokine levels were analyzed. RESULTS Alleviation of inflammation, inhibition of apoptosis, and enhancement of intrinsic MSCs have been established in the SAP-SP group in diverse animal models. Furthermore, the inflammatory effects on human samples were examined in synoviocytes and synovial fluid from patients with OA. In this study, we observed that SAP-SP showed anti-inflammatory action in OA conditions and increased cartilage regeneration by recruiting intrinsic MSCs, inhibiting progression of OA. CONCLUSIONS These therapeutic effects have been validated in diverse OA models, including rabbits, Dunkin Hartley guinea pigs, and human synoviocytes. Therefore, we propose that SAP-SP may be an effective injectable therapeutic agent for treating OA. In this manuscript, we report a preclinical study of novel self-assembled peptide (SAP)-substance P (SP) hydrogels with diverse animal models and human synoviocytes and it displays anti-inflammatory effects, apoptosis inhibition, intrinsic mesenchymal stem cells recruitments and cartilage regeneration.
Collapse
Affiliation(s)
- Sang Jun Kim
- Department of Physical and Rehabilitation Medicine, Seoul Jun Rehabilitation Clinic and Research Center, Seoul, Republic of Korea
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji Eun Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Goeun Choe
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Da Hyun Song
- Department of Physical and Rehabilitation Medicine, Seoul Jun Rehabilitation Clinic and Research Center, Seoul, Republic of Korea
| | - Sun Jeong Kim
- Stem Cell Institute, ENCell Co. Ltd, Seoul, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Tae Hee Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jin Yoo
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Soo Hyun Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Youngmee Jung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
- School of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Little CB. Cholesterol, systemic inflammation, interleukin-1β, and osteoarthritis risk - aligning animal models with specific patient endotypes provides novel insights. Osteoarthritis Cartilage 2023; 31:298-299. [PMID: 36473676 DOI: 10.1016/j.joca.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Affiliation(s)
- C B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales 2065, Australia.
| |
Collapse
|
34
|
Fang Z, Li X, Lei S, Feng S, Zhou C, Tong X, Han R. Protective effects of Pudilan Tablets against osteoarthritis in mice induced by monosodium iodoacetate. Sci Rep 2023; 13:2760. [PMID: 36797333 PMCID: PMC9935914 DOI: 10.1038/s41598-023-29976-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Osteoarthritis (OA) is a complicated disorder that is the most prevalent chronic degenerative joint disease nowadays. Pudilan Tablets (PDL) is a prominent traditional Chinese medicine formula used in clinical settings to treat chronic inflammatory illnesses. However, there is currently minimal fundamental research on PDL in the therapy of joint diseases. As a result, this study looked at the anti-inflammatory and anti-OA properties of PDL in vitro and in vivo, as well as the mechanism of PDL in the treatment of OA. We investigated the anti-OA properties of PDL in OA mice that were generated by monosodium iodoacetate (MIA). All animals were administered PDL (2 g/kg or 4 g/kg) or the positive control drug, indomethacin (150 mg/kg), once daily for a total of 28 days starting on the day of MIA injection. The CCK-8 assay was used to test the vitality of PDL-treated RAW264.7 cells in vitro. RAW264.7 cells that had been activated with lipopolysaccharide (LPS) were used to assess the anti-inflammatory properties of PDL. In the MIA-induced OA model mice, PDL reduced pain, decreased OA-induced cartilage damages and degradation, decreased production of pro-inflammatory cytokines in serum, and suppressed IL-1β, IL-6, and TNF-α mRNA expression levels in tibiofemoral joint. In RAW264.7 cells, PDL treatment prevented LPS-induced activation of the ERK/Akt signaling pathway and significantly decreased the levels of inflammatory cytokines, such as IL-1β, IL-6, and TNF-α. In conclusion, these results suggest that PDL is involved in combating the development and progression of OA, exerts a powerful anti-inflammatory effect on the knee joint, and may be a promising candidate for the treatment of OA.
Collapse
Affiliation(s)
- Zhizheng Fang
- grid.252251.30000 0004 1757 8247School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Xiangyu Li
- Department of Research and Development, Anhui Jiren Pharmaceutical Company, Bozhou, 236800 China
| | - Shujun Lei
- grid.252251.30000 0004 1757 8247School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Shibin Feng
- grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036 China
| | - Chenyu Zhou
- grid.252251.30000 0004 1757 8247School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Xiaohui Tong
- School of Life Sciences, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Rongchun Han
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
35
|
Dou H, Wang S, Hu J, Song J, Zhang C, Wang J, Xiao L. Osteoarthritis models: From animals to tissue engineering. J Tissue Eng 2023; 14:20417314231172584. [PMID: 37223125 PMCID: PMC10201005 DOI: 10.1177/20417314231172584] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/13/2023] [Indexed: 05/25/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative osteoarthropathy. Although it has been revealed that a variety of factors can cause or aggravate the symptoms of OA, the pathogenic mechanisms of OA remain unknown. Reliable OA models that accurately reflect human OA disease are crucial for studies on the pathogenic mechanism of OA and therapeutic drug evaluation. This review first demonstrated the importance of OA models by briefly introducing the OA pathological features and the current limitations in the pathogenesis and treatment of OA. Then, it mainly discusses the development of different OA models, including animal and engineered models, highlighting their advantages and disadvantages from the perspective of pathogenesis and pathology analysis. In particular, the state-of-the-art engineered models and their potential were emphasized, as they may represent the future direction in the development of OA models. Finally, the challenges in obtaining reliable OA models are also discussed, and possible future directions are outlined to shed some light on this area.
Collapse
Affiliation(s)
- Hongyuan Dou
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Shuhan Wang
- Shenzhen Institute for Drug Control, Shenzhen Testing Center of Medical Devices, Shenzhen, China
| | - Jiawei Hu
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Jian Song
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Jiali Wang
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
36
|
Wang S, Ma J, Zhao X, Xue Y, Liu W, Huang H, Zhang L, Tian A, Ma X. The Osteoarthritis Natural Progress and Changes in Intraosseous Pressure of the Guinea Pig Model in Different Degeneration Stages. Orthop Surg 2022; 14:3036-3046. [PMID: 36168980 DOI: 10.1111/os.13496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Articular cartilage and subchondral bone changes during the pathological progress of knee osteoarthritis (KOA) is a key event marking the development of the disease. The age varying alteration patterns within entire osteochondral unit remains poorly understood. The purpose of this study was to find a reasonable age range of the Dunkin-Hartley guinea pig model for the studying of KOA pathological process, and to investigate Intraosseous pressure (IOP) in the process during different degeneration stages of KOA. METHODS Male Dunkin-Hartley guinea pigs were selected and divided into groups of 3, 6, 9, 12, 18 months old by age, 10 in each group. All knees underwent imaging examination including X-ray, Micro-CT and MRI. Observed the imaging findings with the use of Kellgren-Lawrence (K-L) classification and knee osteoarthritis MRI scores. Measured the IOP of distal femur (DF) and proximal tibia (PT) in each group, and observed the differences of bilateral tibiofemoral articular cartilage in histological and immunohistochemistry, staining results were evaluated by using Mankin's score. Analysis of variance (ANOVA) and t-tests were used to compare the differences indicators between groups. RESULTS With the increase of age, changes in X-ray, Micro-CT and MRI imaging findings and pathological staining results of articular cartilage in all stages were consistent with the changing of degenerative KOA process. The IOP of DF and PT increased gradually with age, and reached its peak in 12-month age group, and then gradually decreased, there was a statistically significant difference of IOP between each group. The IOP of DF was slightly higher than that of PT, but the difference was not statistically significant. CONCLUSION Dunkin-Hartley guinea pigs can be used as an animal model to study different pathological stages of KOA. There might be a correlation between the changes of IOP and the pathological progress of articular cartilage and subchondral bone in DF and PT.
Collapse
Affiliation(s)
- Shuo Wang
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin University, Tianjin, China.,Department of Orthopaedics, Chinese People's Liberation Army Joint Logistics Support Force Tianjin Rehabilitation Center (Former No. 464 Hospital of People's Liberation Army), Tianjin, China
| | - Jianxiong Ma
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Xingwen Zhao
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Yuan Xue
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Liu
- Department of Orthopaedics, Tianjin Baodi Hospital, Tianjin, China
| | - Hongchao Huang
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Lei Zhang
- Department of Orthopaedics, Tianjin Medical University Hospital for Metabolic Diseases, Tianjin, China
| | - Aixian Tian
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Xinlong Ma
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
37
|
Little CB, Zaki S, Blaker CL, Clarke EC. Animal models of osteoarthritis. Bone Joint Res 2022; 11:514-517. [PMID: 35909339 PMCID: PMC9396918 DOI: 10.1302/2046-3758.118.bjr-2022-0217.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Cite this article: Bone Joint Res 2022;11(8):514–517.
Collapse
Affiliation(s)
- Christopher B. Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
| | - Sanaa Zaki
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Sydney, Australia
| | - Carina L. Blaker
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
| | - Elizabeth C. Clarke
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
| |
Collapse
|
38
|
Liu Y, Liu J, Ma Y, Zhang Y, Chen Q, Yang X, Shang Y. The protective effects of Olmesartan against interleukin-29 (IL-29)-induced type 2 collagen degradation in human chondrocytes. Bioengineered 2022; 13:1802-1813. [PMID: 35012432 PMCID: PMC8805962 DOI: 10.1080/21655979.2021.1997090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/19/2021] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is a cartilage degenerative disease commonly observed in the elderly population and is pathologically characterized by the degradation of the cartilage extracellular matrix (ECM). Matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs) are critical enzymes involved in the degradation of ECM. Olmesartan is an inhibitor of the angiotensin II receptor developed for the treatment of hypertension, and recent studies show that it exerts anti-inflammatory effects in arthritis. The present study aimed to investigate the mechanism of the protective effect of Olmesartan on cartilage ECM degradation. Interleukin-29 (IL-29) is a novel inflammatory mediator involved in the inflammation and degradation of cartilage in OA, and human T/C-28a2 cells treated with it were the inflammatory model in vitro. We found that the degradation of type 2 collagens and aggrecans was induced by IL-29, accompanied by the upregulation of MMPs and ADAMTSs, but the presence of Olmesartan significantly ameliorated these increases. In addition, Olmesartan abolished IL-29- induced oxidative stress and elevated the expression level of TNF receptor-associated factor 6 (TRAF-6). Mechanistically, we showed that Olmesartan suppressed IL-29- caused inhibitor kappa B α (IκBα) expression and nuclear translocation of nuclear factor kappa-B (NF-κB) p65, indicating it suppressed the activation of the NF-κB pathway. Collectively, our data reveal that Olmesartan exerted a protective function on IL-29- induced type 2 collagen degradation in human chondrocytes.
Collapse
Affiliation(s)
- Yunlong Liu
- Department of Knee Surgery, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Junyi Liu
- Department of Knee Surgery, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Yan Ma
- Lab of Molecular Biology, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Yongyong Zhang
- Lab of Molecular Biology, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Qiong Chen
- Department of Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xin Yang
- Department of Knee Surgery, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Yanchun Shang
- Department of Knee Surgery, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| |
Collapse
|
39
|
Haubruck P, Pinto MM, Moradi B, Little CB, Gentek R. Monocytes, Macrophages, and Their Potential Niches in Synovial Joints - Therapeutic Targets in Post-Traumatic Osteoarthritis? Front Immunol 2021; 12:763702. [PMID: 34804052 PMCID: PMC8600114 DOI: 10.3389/fimmu.2021.763702] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Synovial joints are complex structures that enable normal locomotion. Following injury, they undergo a series of changes, including a prevalent inflammatory response. This increases the risk for development of osteoarthritis (OA), the most common joint disorder. In healthy joints, macrophages are the predominant immune cells. They regulate bone turnover, constantly scavenge debris from the joint cavity and, together with synovial fibroblasts, form a protective barrier. Macrophages thus work in concert with the non-hematopoietic stroma. In turn, the stroma provides a scaffold as well as molecular signals for macrophage survival and functional imprinting: “a macrophage niche”. These intricate cellular interactions are susceptible to perturbations like those induced by joint injury. With this review, we explore how the concepts of local tissue niches apply to synovial joints. We introduce the joint micro-anatomy and cellular players, and discuss their potential interactions in healthy joints, with an emphasis on molecular cues underlying their crosstalk and relevance to joint functionality. We then consider how these interactions are perturbed by joint injury and how they may contribute to OA pathogenesis. We conclude by discussing how understanding these changes might help identify novel therapeutic avenues with the potential of restoring joint function and reducing post-traumatic OA risk.
Collapse
Affiliation(s)
- Patrick Haubruck
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Marlene Magalhaes Pinto
- Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Babak Moradi
- Clinic of Orthopaedics and Trauma Surgery, University Clinic of Schleswig-Holstein, Kiel, Germany
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Rebecca Gentek
- Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|