1
|
Wang J, Sun L, Zhang Y, Chen S, He Y. Wnt/β-catenin regulates Gli1 + osteogenic progenitors in condylar subchondral bone development and osteoarthritis. BMC Musculoskelet Disord 2025; 26:533. [PMID: 40448062 DOI: 10.1186/s12891-025-08765-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 05/15/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Gli1 has been identified as a marker of osteogenic progenitors in the condylar subchondral bone. The Wnt/β-catenin signaling pathway is known to regulate stem cell proliferation and differentiation in bone. Whether Wnt/β-catenin signaling pathway could influence Gli1 + osteogenic progenitors remains unclear. Here, we aimed to investigate the role and related mechanisms of Wnt/β-catenin signaling in the regulation of Gli1 + osteogenic progenitors in condylar development and temporomandibular joint osteoarthritis (TMJOA). METHODS We generated Gli1-CreERT2;tdTomato mice to perform lineage tracing; We generated Gli1-CreERT2; β-cateninfl/fl mice, in which β-catenin was lost in the Gli1 + lineage to examine the role of Wnt/β-catenin signaling pathway in regulating the proliferation and differentiation of Gli1 + cells. The β-catenin CKO mice and their wild-type (WT) littermates were induced at 3 days and were euthanized 1, 2 or 4 weeks after induction; We induced a TMJOA model through a unilateral partial discectomy (UPD) of the temporomandibular joint disc in 6-week-old tamoxifen-treated Gli1-CreERT2;β-cateninfl/fl;tdTomato mice and control group (Gli1-CreERT2;tdTomato mice). We harvested the mandibles at 4 weeks post-surgery. RESULTS Conditional knockout of β-catenin inhibited the osteogenic activity of Gli1 + progenitor cells during condylar subchondral bone development. In discectomy-induced TMJOA, the overactivation of Gli1 in subchondral bone drove pathological osteogenesis and aberrant subchondral bone remodeling. Deletion of β-catenin in Gli1 + cells mitigated excessive Gli1 + cells activation and ectopic mineralization. CONCLUSION Our findings establish Wnt/β-catenin signaling as a key regulator of Gli1 + progenitor cell fate determination in both bone development and TMJOA pathogenesis.
Collapse
Affiliation(s)
- Jie Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, P. R. China
| | - Lin Sun
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, P. R. China
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, P. R. China
| | - Shuo Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, P. R. China.
| | - Yang He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, P. R. China.
| |
Collapse
|
2
|
Zhou Y, Yu S, Xue B, Zhang X, Gao M, Liu Y. Analysis and ceRNA Network Construction of Differentially Expressed lncRNAs and mRNAs in Human Osteoarthritis Cartilage. Biochem Genet 2025:10.1007/s10528-025-11131-1. [PMID: 40358893 DOI: 10.1007/s10528-025-11131-1] [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: 11/15/2024] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
This study aimed to identify differentially expressed long non-coding RNAs (lncRNAs) and messenger RNAs (mRNAs) in damaged cartilage (DC) and un-damaged cartilage (UDC) in human osteoarthritis (OA), exploring their roles in disease progression through bioinformatics analysis and ceRNA network construction. Cartilage samples from 5 OA patients undergoing total knee arthroplasty were analyzed. RNA sequencing was used to detect the expression of lncRNAs and mRNAs in DC and UDC samples. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to investigate biological processes. A ceRNA network was constructed, and differentially expressed RNAs were validated by quantitative real-time polymerase chain reaction (qRT-PCR). In the damaged cartilage (DC) samples, 5 lncRNAs were significantly upregulated, and 15 were significantly downregulated, while 8 mRNAs were upregulated, and 8 were downregulated. The differential expression of lncRNAs, including LINC01411, AL596087.2, PCDH20, LRFN2, and AL583785.1, was confirmed using qRT-PCR, with p-values for all results showing statistical significance (p < 0.05). GO/KEGG enrichment analysis revealed key pathways such as Ras, PI3K-Akt, and MAPK that were significantly involved in OA pathogenesis. The ceRNA network construction highlighted crucial miRNA interactions, identifying potential regulators of cartilage-related biological processes. Differentially expressed lncRNAs and mRNAs are involved in critical signaling pathways in OA cartilage, suggesting their potential as biomarkers or therapeutic targets for OA treatment. Further functional studies are needed to fully elucidate their roles in OA pathogenesis.
Collapse
Affiliation(s)
- Yong Zhou
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Shengyuan Yu
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Bing Xue
- The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xi Zhang
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ming Gao
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yishu Liu
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Rahaman SN, Lishadevi M, Anandasadagopan SK. Unraveling the Molecular Mechanisms of Osteoarthritis: The Potential of Polyphenols as Therapeutic Agents. Phytother Res 2025; 39:2038-2071. [PMID: 40044420 DOI: 10.1002/ptr.8455] [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: 06/27/2024] [Revised: 01/17/2025] [Accepted: 01/25/2025] [Indexed: 05/21/2025]
Abstract
The complex nature of osteoarthritis (OA), driven by the intricate interplay of genetic, environmental, and lifestyle factors, necessitates the development of a single treatment method, which is highly challenging. The long-term use of non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids often leads to adverse side effects like kidney damage and stomach ulcers. Major health threats like obesity and aging create a milieu of chronic low-grade inflammation and increased mechanical stress on the joints resulting in cartilage deterioration. Additionally, postmenopausal women with lower circulating 17β-estradiol levels experience accelerated joint deterioration due to increased immune activity resulting in the increased production of pro-inflammatory cytokines, with elevated MMP expression and decreased type II collagen synthesis. Polyphenols are nature's gifted magic molecules, which possess diverse biological properties like anti-oxidant, anti-bacterial, anti-inflammatory, estrogenic, and insulin-sensitizing effects, which can manage and treat all the multi-factorial contributing factors of OA effectively. Certain polyphenols can act as phytoestrogens and mimic the effects of natural estrogen by binding to ERα and ERβ and can act as SERMs and prevent degradation of the articular cartilage thereby alleviating osteoarthritic conditions. These molecules downregulate the expression of various pro-inflammatory cytokines, apoptotic genes, and matrix-degrading proteases (MMPs) while upregulating major ECM proteins like type II collagen, aggrecan, and proteoglycans in various osteoarthritic animal models. This review provides a comprehensive overview of the molecular mechanisms involved in OA development and also explores the therapeutic potential of different polyphenols in mitigating joint inflammation and their protective effect in inhibiting the degradation of cartilage extracellular matrix (ECM) and enhancing joint homeostasis.
Collapse
Affiliation(s)
- Syed Nasar Rahaman
- Biochemistry and Biotechnology Laboratory, Central Leather Research Institute, Council of Scientific and Industrial Research (CSIR), Chennai, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Murugesan Lishadevi
- Biochemistry and Biotechnology Laboratory, Central Leather Research Institute, Council of Scientific and Industrial Research (CSIR), Chennai, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Suresh Kumar Anandasadagopan
- Biochemistry and Biotechnology Laboratory, Central Leather Research Institute, Council of Scientific and Industrial Research (CSIR), Chennai, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
4
|
Łabędź-Masłowska A, Wieczorek J, Mierzwiński M, Sekuła-Stryjewska M, Noga S, Rajca J, Duda P, Milian-Ciesielska K, Karnas E, Kmiotek-Caller K, Szkaradek A, Madeja Z, Ficek K, Jura J, Zuba-Surma E. Evaluation of the Safety and Regenerative Potential of Human Mesenchymal Stem Cells and Their Extracellular Vesicles in a Transgenic Pig Model of Cartilage-Bone Injury In Vivo - Preclinical Study. Stem Cell Rev Rep 2025; 21:1075-1095. [PMID: 40380984 PMCID: PMC12102096 DOI: 10.1007/s12015-025-10853-4] [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] [Accepted: 02/07/2025] [Indexed: 05/19/2025]
Abstract
Osteoarthritis (OA) is a degenerative joint condition leading to disability. The lack of effective treatment for OA creates a need for the development of new therapeutic approaches that may rely on stem cells including mesenchymal stem/stromal cells (MSCs) and their derivatives such as extracellular vesicles (EVs). The objective of this study was to evaluate the impact of MSCs derived from adipose tissue (AT-MSCs) and umbilical cord (UC-MSCs) and their EVs on cartilage-bone injury in vivo, to identify the specimen with the highest regenerative potential for further clinical applications in patients with OA. Humanized pigs underwent cartilage-bone injuries followed by intraarticular administration of products containing AT-MSCs, UC-MSCs, AT-MSC-EVs or UC-MSC-EVs mixed with hyaluronic acid (HA) or HA alone (for comparison). After 6-m follow-up, almost-fully-healed cartilage-bone defects were observed in the AT-MSC- and UC-MSC-treated pigs, and the defects were filled primarily with hyaline cartilage. In AT-MSC-EV- and UC-MSC-EV-treated pigs, a partial cartilage-bone tissue repair was observed, and the defects were filled primarily with fibrocartilage. The control pigs demonstrated limited regeneration capacity. The microcomputed tomography parameters of the subchondral bone indicated the ongoing progression of OA in controls, whereas in the MSC- and MSC-EV-treated pigs, the parameters indicated the cessation of OA progression. Moreover, no serious side effects were observed after the administration of products containing MSCs or MSC-EVs. The results indicate the safety and regenerative activity of MSCs on injured tissues, which favors not only the healing and improvement of bone structure but also the formation of hyaline cartilage. Superior tissue repair was observed after the administration of products containing AT-MSCs. The treatment of OA with MSC-EVs needs further standardization.
Collapse
Affiliation(s)
- Anna Łabędź-Masłowska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jarosław Wieczorek
- University Center of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, Krakow, Poland
| | - Maciej Mierzwiński
- Department of Science, Innovation and Development, Galen-Orthopaedics, Bierun, Poland
| | - Małgorzata Sekuła-Stryjewska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Sylwia Noga
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Laboratory of Stem Cell Biotechnology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jolanta Rajca
- Department of Science, Innovation and Development, Galen-Orthopaedics, Bierun, Poland
- Spin-Lab Centre for Microscopic Research on Matter, University of Silesia in Katowice, Katowice, Poland
| | - Piotr Duda
- Institute of Biomedical Engineering, Faculty of Science and Technology, University of Silesia in Katowice, Katowice, Poland
| | | | - Elżbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Katarzyna Kmiotek-Caller
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Agnieszka Szkaradek
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Krzysztof Ficek
- Department of Science, Innovation and Development, Galen-Orthopaedics, Bierun, Poland
| | - Jacek Jura
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Balice, Poland.
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
5
|
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
|
6
|
Zhou Y, Li M, Lin S, Zhu Z, Zhuang Z, Cui S, Chen L, Zhang R, Wang X, Shen B, Chen C, Yang R. Mechanical sensing protein PIEZO1 controls osteoarthritis via glycolysis mediated mesenchymal stem cells-Th17 cells crosstalk. Cell Death Dis 2025; 16:231. [PMID: 40169556 PMCID: PMC11961634 DOI: 10.1038/s41419-025-07577-1] [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: 07/30/2024] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025]
Abstract
Aberrant mechanical stimuli can cause tissue attrition and activate mechanosensitive intracellular signaling, impacting the progression of osteoarthritis (OA). However, the precise relationship between mechanical loading and bone metabolism remains unclear. Here, we present evidence that Piezo1 senses the mechanical stimuli to coordinate the crosstalk between mesenchymal stem cells (MSCs) and T helper 17 (Th17) cells, leading to the deterioration of bone and cartilage in osteoarthritis (OA). Mechanical loading impaired the property of MSCs by inhibiting their osteo-chondrogenic differentiation and promoting inflammatory signaling to enhance Th17 cells. Mechanistically, mechanical stimuli activated Piezo1, thereby facilitating Ca2+ influx which upregulated the activity of Hexokinase 2(HK2), the rate-limiting enzyme of glycolysis. The resultant increase in glycolytic activity enhanced communication between MSCs and T cells, thus promoting Th17 cell polarization in a macrophage migration inhibitory factor (MIF) dependent manner. Functionally, Wnt1cre; Piezo1fl/fl mice reduced bone and cartilage erosion in the temporomandibular joint condyle following mechanical loading compared to control groups. Additionally, we observed activated Piezo1 and HK2-mediated glycolysis in patients with temporomandibular joint OA, thereby confirming the clinical relevance of our findings. Overall, our results provide insights into how Piezo1 in MSCs coordinates with mechano-inflammatory signaling to regulate bone metabolism. The schema shows that mechanical sensing protein PIEZO1 in MSCs controls osteoarthritis via glycolysis mediated MSCs and Th17 cells crosstalk in a MIF dependent manner.
Collapse
Affiliation(s)
- Yikun Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
- Department of Orthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mingzhao Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Shuai Lin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Zilu Zhu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Zimeng Zhuang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Shengjie Cui
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Liujing Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Ran Zhang
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xuedong Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Bo Shen
- National Institute of Biological Sciences, Beijing, China, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Ruili Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China.
| |
Collapse
|
7
|
Shin Y, Kwak JS, Kim SK, Chun JS. Fibroblast growth factor 7 (FGF7) causes cartilage destruction, subchondral bone remodeling, and the premature growth plate closure in mice. Osteoarthritis Cartilage 2025; 33:426-436. [PMID: 39638118 DOI: 10.1016/j.joca.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Fibroblast growth factor (FGF) signaling plays a significant role in osteoarthritis (OA) pathogenesis, though the OA-related functions of only a few FGFs have been fully elucidated. This study investigates the specific roles of FGF7 in OA development. METHODS FGF7 expression was analyzed in human (n=6) and mouse (n=10) cartilage. Experimental OA was induced by destabilization of the medial meniscus (DMM). The roles of FGF7 were explored using intra-articular (IA) injection of recombinant FGF7 (rFGF7) and whole-body Fgf7 knockout mice (Fgf7-/-). Subchondral bone remodeling and growth plate morphology were assessed micro computed tomography (µCT) and histological analysis. RESULTS FGF7 was upregulated in OA cartilage. IA injection of rFGF7 led to OA cartilage destruction (OARSI [Osteoarthritis Research Society International] grade; 0.61 [95% CI 0.00-5.33]), while Fgf7-/- mice showed reduced DMM-induced cartilage erosion (OARSI grade; 1.89 [95% CI 1.08-3.00]) compared to wild-type mice (4.92 [95% CI 3.83-5.33]). These effects were associated with changes in matrix-degrading enzyme expression in chondrocytes. Mice receiving IA injection of rFGF7 (20 μg) exhibited increased subchondral bone thickness (68.01 µm [95% CI 61.55-74.46]) and decreased osteoclastogenesis (tartrate-resistant acid phosphatase positivity; 1.94% [95% CI 1.41-2.47]) compared to controls (38.33 µm [95% CI 33.71-42.96]) and (4.23% [95% CI 3.28-5.19]), respectively. Additionally, rFGF7 treatment caused premature closure of growth plates, whereas Fgf7-/- mice exhibited significantly increased growth plate thickness. CONCLUSIONS FGF7 exerts multiple functions in various joint tissues, including promoting cartilage destruction, inducing subchondral bone remodeling (SBP thickening), and triggering premature growth plate closure.
Collapse
MESH Headings
- Animals
- Bone Remodeling/drug effects
- Bone Remodeling/physiology
- Growth Plate/pathology
- Growth Plate/metabolism
- Growth Plate/drug effects
- Growth Plate/diagnostic imaging
- Fibroblast Growth Factor 7/pharmacology
- Fibroblast Growth Factor 7/metabolism
- Fibroblast Growth Factor 7/genetics
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cartilage, Articular/drug effects
- Cartilage, Articular/diagnostic imaging
- Mice
- Mice, Knockout
- Humans
- X-Ray Microtomography
- Male
- Chondrocytes/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Experimental/metabolism
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/pathology
- Middle Aged
- Female
- Injections, Intra-Articular
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Youngnim Shin
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Ji-Sun Kwak
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Seul Ki Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jang-Soo Chun
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
8
|
Hashimoto T, Akagi M, Tsukamoto I, Hashimoto K, Morishita T, Ito T, Goto K. RANKL-mediated osteoclastic subchondral bone loss at a very early stage precedes subsequent cartilage degeneration and uncoupled bone remodeling in a mouse knee osteoarthritis model. J Orthop Surg Res 2025; 20:226. [PMID: 40025588 PMCID: PMC11874437 DOI: 10.1186/s13018-025-05578-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION Uncoupled bone remodeling in the subchondral bone (SB) has recently been considered as an important process in the progression of knee osteoarthritis (KOA). In this study, we aimed to investigate changes in SB and articular cartilage using a mouse model of destabilization of the medial meniscus (DMM) and determine the effects of bone metabolism on KOA progression. METHODS DMM or sham surgery was performed on the left knees of 40-week-old male wild-type (WT) mice and Tsukuba hypertensive mice (THM), which exhibit high-turnover bone metabolism. Bone volume/tissue volume (BV/TV) and bone mineral density (BMD) in the medial tibial SB were measured longitudinally in vivo using μCT at 0 (immediately after surgery), 1, 2, 4, 8, and 12 weeks postoperatively. Concurrently, histological evaluations of the articular cartilage in the medial tibial plateau were conducted. Furthermore, the number of endo-periosteal tartrate-resistant acid phosphatase-positive osteoclasts, trabecular RANKL-positive osteocytes, and osteocytes in the trabeculae were measured at 0, 1, 2, and 4 weeks. RESULTS In the WT + DMM group, BV/TV and BMD in the SB significantly decreased with time, whereas cartilage degeneration significantly increased. In the THM + DMM group, these changes in BMD and cartilage degeneration were significantly pronounced. Interestingly, in the THM + DMM group, BV/TV significantly decreased up to 4 weeks but then began to increase, although BMD continued to decrease until the 12-week mark. The number of osteoclasts and the percentage of RANKL-positive osteocytes per total number of osteocytes within the total trabecular bone area (%) in the WT + DMM group significantly increased with time, with a significant difference between the WT + DMM and WT + sham groups at 4 weeks. The number of osteocytes in the WT + DMM group significantly decreased with time, and the difference between the WT + DMM and WT + sham groups was significant at 4 weeks postoperatively. These histological changes were significantly enhanced in the THM + DMM group. CONCLUSIONS The results indicate that early-stage osteocyte death in the SB and RANKL-mediated osteoclastic SB loss precede histological cartilage degeneration and contribute to uncoupled bone remodeling at the later stage. Acceleration of disease processes in the THM + DMM group suggests that high-turnover bone metabolism is a potential risk factor for KOA. Maintaining SB integrity and avoiding continuous SB overload may be key strategies for mitigating disease progression.
Collapse
Affiliation(s)
- Teruaki Hashimoto
- Department of Orthopaedic Surgery, Kindai University Hospital, 377-2 Ohno-Higashi, Osaka-Sayama City, Osaka, 589-8511, Japan.
| | - Masao Akagi
- Department of Orthopaedic Surgery, Kindai University Hospital, 377-2 Ohno-Higashi, Osaka-Sayama City, Osaka, 589-8511, Japan
| | - Ichiro Tsukamoto
- Department of Orthopaedic Surgery, Kindai University Hospital, 377-2 Ohno-Higashi, Osaka-Sayama City, Osaka, 589-8511, Japan
| | - Kazuhiko Hashimoto
- Department of Orthopaedic Surgery, Kindai University Hospital, 377-2 Ohno-Higashi, Osaka-Sayama City, Osaka, 589-8511, Japan
| | - Takafumi Morishita
- Department of Orthopaedic Surgery, Kindai University Hospital, 377-2 Ohno-Higashi, Osaka-Sayama City, Osaka, 589-8511, Japan
| | - Tomohiko Ito
- Department of Orthopaedic Surgery, Kindai University Hospital, 377-2 Ohno-Higashi, Osaka-Sayama City, Osaka, 589-8511, Japan
| | - Koji Goto
- Department of Orthopaedic Surgery, Kindai University Hospital, 377-2 Ohno-Higashi, Osaka-Sayama City, Osaka, 589-8511, Japan
| |
Collapse
|
9
|
Wang R, Mehrjou B, Dehghan‐Banian D, Wang BYH, Li Q, Deng S, Liu C, Zhang Z, Zhu Y, Wang H, Li D, Lu X, Cheng JCY, Ong MTY, Chan HF, Li G, Chu PK, Lee WYW. Targeting Long Noncoding RNA H19 in Subchondral Bone Osteocytes and the Alleviation of Cartilage Degradation in Osteoarthritis. Arthritis Rheumatol 2025; 77:283-297. [PMID: 39482250 PMCID: PMC11865692 DOI: 10.1002/art.43028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVE Emerging evidence suggests long noncoding RNA H19 is associated with osteoarthritis (OA) pathology. However, how H19 contributes to OA has not been reported. This study aims to investigate the biologic function of H19 in OA subchondral bone remodeling and OA progression. METHODS Clinical joint samples and OA animal models induced by surgical destabilization of the medial meniscus (DMM) were used to verify the causal relationship between osteocyte H19 and OA subchondral bone and cartilage changes. MLO-Y4 osteocyte cells subjected to fluid shear stress were used to verify the mechanism underlying H19-mediated mechanoresponse. Finally, the antisense oligonucleotide (ASO) against H19 was delivered to mice knee joints by magnetic metal-organic framework (MMOF) nanoparticles to develop a site-specific delivery method for targeting osteocyte H19 for OA treatment. RESULTS Both clinical OA subchondral bone and wildtype mice with DMM-induced OA exhibit aberrant higher subchondral bone mass, with more H19 mice expressing osteocytes. On the contrary, mice with osteocyte-specific deletion of H19 are less vulnerable to DMM-induced OA phenotype. In MLO-Y4 cells, H19-mediated osteocyte mechanoresponse through PI3K/AKT/GSK3 signal activation by EZH2-induced H3K27me3 regulation on protein phosphatase 2A inhibition. Targeted inhibition of H19 (using ASO-loaded MMOF) substantially alleviates subchondral bone remodeling and OA phenotype. CONCLUSION In summary, our results provide new evidence that the elevated H19 expression in osteocytes may contribute to aberrant subchondral bone remodeling and OA progression. H19 appears to be required for the osteocyte response to mechanical stimulation, and targeting H19 represents a new promising approach for OA treatment.
Collapse
Grants
- 2020 Rising Star Award, American Society for Bone and Mineral Research
- AoE/M-402/20 Area of Excellence, University Grants Committee, Hong Kong SAR
- Start-up Fund, The Chinese University of Hong Kong, Hong Kong SAR
- Matching Grant Scheme, University Grants Committee, Hong Kong SAR
- 2412162 General Research Fund, University Grants Committee, Hong Kong SAR
- CT1.1 Center for Neuromusculoskeletal Restorative Medicine, Health@InnoHK Program, Innovation Technology Commission, Hong Kong SAR
Collapse
Affiliation(s)
- Rongliang Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Babak Mehrjou
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong Kong SARChina
| | - Dorsa Dehghan‐Banian
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Belle Yu Hsuan Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Center for Neuromusculoskeletal Restorative MedicineCUHK InnoHK Centres, Hong Kong Science ParkHong Kong SARChina
| | - Qiangqiang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Shuai Deng
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Chuanhai Liu
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Zhe Zhang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and SH Ho Scoliosis Research LaboratoryJoint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, ShatinHong Kong SARChina
| | - Yanlun Zhu
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Haixing Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Dan Li
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong Kong SARChina
| | - Xiaomin Lu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Jack Chun Yiu Cheng
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and SH Ho Scoliosis Research LaboratoryJoint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, ShatinHong Kong SARChina
| | - Michael Tim Yun Ong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales HospitalThe Chinese University of Hong Kong, ShatinHong Kong SARChina
| | - Hon Fai Chan
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Paul K. Chu
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong Kong SARChina
| | - Wayne Yuk Wai Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science ParkHong Kong SARChina
| |
Collapse
|
10
|
Feng SY, Cao MN, Gao CC, Li YX, Lei J, Fu KY. Akt2 inhibition alleviates temporomandibular joint osteoarthritis by preventing subchondral bone loss. Arthritis Res Ther 2025; 27:43. [PMID: 40016746 PMCID: PMC11866854 DOI: 10.1186/s13075-025-03506-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND This study aimed to investigate the role and mechanism of the Akt2 pathway in different stages of anterior disc displacement (ADD)-induced temporomandibular joint osteoarthritis (TMJOA). METHODS A rat model for TMJOA that simulates anterior disc displacement was established. For inhibit Akt2 expression in subchondral bone, rats were intravenously injected with adeno-associated virus carrying Akt2 shRNA at a titer of 1 × 1012 transducing units/mL 10 days before the ADD or sham operations. The rats were euthanized and evaluated 1 or 8 weeks after surgery, as these time points represented the early or advanced stage of ADD. Immunostaining was performed to examine the expression and location of phosphorylated Akt2 in different stages of ADD. Microcomputed tomography, hematoxylin and eosin staining, toluidine blue staining, Western blotting, immunohistochemical and immunofluorescence staining were used to elucidate the pathological changes and potential mechanisms underlying ADD-induced TMJOA. RESULTS In the rat model of ADD-induced TMJOA, rapid condylar bone loss occurred with increased phosphorylation of Akt2 in subchondral bone macrophages within 1 week post-surgery. At 8 weeks after surgery, abnormal remodeling of subchondral bone and degenerative changes in cartilage were observed. Inhibiting Akt2 reduced condylar bone resorption following ADD surgery while improving condylar bone morphology at 8 weeks post-surgery. Additionally, inhibition of Akt2 alleviated cartilage degeneration characterized by a decreased number of apoptotic chondrocytes, reduced expression of matrix metalloproteinases, and increased collagen type II expression in cartilage tissue. CONCLUSIONS The Akt2 pathway is activated mainly in subchondral bone macrophages during the early stage of ADD and plays an important role in regulating subchondral bone remodeling. Inhibition of Akt2 could serve as a prophylactic treatment to slow the progression of ADD-induced TMJOA.
Collapse
Affiliation(s)
- Shi-Yang Feng
- Center for TMD & Orofacial Pain, Peking University School and Hospital of Stomatology, No. 22 Zhong Guan Cun South Ave, Beijing, 100081, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Meng-Nan Cao
- Center for TMD & Orofacial Pain, Peking University School and Hospital of Stomatology, No. 22 Zhong Guan Cun South Ave, Beijing, 100081, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Chen-Chen Gao
- Center for TMD & Orofacial Pain, Peking University School and Hospital of Stomatology, No. 22 Zhong Guan Cun South Ave, Beijing, 100081, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yi-Xin Li
- Center for TMD & Orofacial Pain, Peking University School and Hospital of Stomatology, No. 22 Zhong Guan Cun South Ave, Beijing, 100081, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Jie Lei
- Center for TMD & Orofacial Pain, Peking University School and Hospital of Stomatology, No. 22 Zhong Guan Cun South Ave, Beijing, 100081, China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China.
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
- Beijing Key Laboratory of Digital Stomatology, Beijing, China.
| | - Kai-Yuan Fu
- Center for TMD & Orofacial Pain, Peking University School and Hospital of Stomatology, No. 22 Zhong Guan Cun South Ave, Beijing, 100081, China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases, Beijing, China.
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
- Beijing Key Laboratory of Digital Stomatology, Beijing, China.
| |
Collapse
|
11
|
Zhu Y, Zhu J, Wang X, Wang P, Liu R. Molecular roles in membrane receptor signaling pathways and cascade reactions in chondrocytes: a review. J Mol Histol 2025; 56:94. [PMID: 39988650 DOI: 10.1007/s10735-025-10368-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/03/2025] [Indexed: 02/25/2025]
Abstract
Articular cartilage (AC) is a specialized connective tissue with unique biological and mechanical properties, which depends on the biological effects of each resident chondrocyte and its surrounding extracellular matrix (ECM) to form a unit that operates in a constant and balanced feedback loop. The surface membrane receptors of chondrocytes play a crucial role in the feedback balance of this biological unit. Various biological signals outside chondrocytes, such as water-soluble chemical signal molecules and mechanical signals, are unable to directly enter the cell and must first bind to the plasma membrane receptors to induce changes in the level and activity of intracellular signal transduction molecules. These changes then transmit through signaling cascade pathways into the nucleus, changing the cell phenotype, and producing physiological or pathological changes. Specific chemical and mechanical signals break the feedback balance of cartilage tissue units through membrane receptors. In the ECM environment, the molecular actions of chondrocyte membrane receptors in response to these specific signals, along with associated ion channel receptors, collectively regulate the biological effects of chondrocytes. This leads to decreased chondrocyte survival and an imbalance in ECM regulation, ultimately disrupting the tissue's molecular framework and physiological feedback mechanisms, and resulting in pathological changes in cartilage tissue. To provide insights into addressing the complexities associated with cartilage tissue injury and repair engineering, this review provides a comprehensive overview of the molecular mechanisms and biological implications of chondrocyte membrane receptor-mediated signal transduction, including G protein-coupled receptors (GPCRs), enzyme-linked receptors (tyrosine kinase receptors (TKRs)), and integrin receptors.
Collapse
Affiliation(s)
- Yingkang Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jingjing Zhu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xu Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Pengbo Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ruiyu Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
12
|
Kim MJ, Yang YJ, Heo JW, Son JD, You YZ, Yang JH, Park KI. Potential Chondroprotective Effect of Artemisia annua L. Water Extract on SW1353 Cell. Int J Mol Sci 2025; 26:1901. [PMID: 40076528 PMCID: PMC11899987 DOI: 10.3390/ijms26051901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Inflammation plays a critical role in the pathogenesis of osteoarthritis (OA). The objective of this study was to investigate the anti-inflammatory and chondroprotective properties of Artemisia annua L. water extract (AWE) following the induction of inflammation in cartilage cells (SW1353 cell) through the administration of interleukin-1 beta (IL-1β). We demonstrated significant antioxidant activity, as evidenced by elevated total phenolic and flavonoid content, in addition to robust free radical scavenging capacity, as assessed through DPPH (2,2-diphenyl-1-picrylhydrazyl) and ABTS (2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) assays. Its cytotoxic effects were assessed at a concentration of 200 μg/mL, where no cytotoxic signs were observed in SW1353 cells treated with IL-1β; the levels of reactive oxygen species (ROS) were notably reduced in a dose-dependent manner. The principal inflammatory markers, cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS), were significantly diminished by AWE treatment. AWE administration led to a dose-dependent reduction in the expression of key proteins involved in the mitogen-activated protein kinase (MAPK) and nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) signaling pathways, ultimately resulting in a decrease in the release of matrix metalloproteinases (MMPs), specifically MMP-1 and MMP-13, which are known to contribute to cartilage degradation. Additionally, the levels of degraded collagen type II in the cartilage cells were restored. These findings suggest that reducing oxidative stress and inflammation, along with inhibiting activated MAPK and NF-κB signaling pathways, may ameliorate the progression of IL-1β-induced OA. Furthermore, a molecular docking analysis revealed a strong binding affinity of MMP-13, a critical mediator in the pathogenesis of OA. Six compounds were identified in AWE, corroborating its potential antioxidant and anti-inflammatory effects. Therefore, AWE may serve as a potentially useful therapeutic agent against OA by modulating inflammation-related mechanisms.
Collapse
Affiliation(s)
- Min Jung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea; (M.J.K.); (Y.J.Y.); (J.W.H.); (J.-d.S.); (Y.Z.Y.)
| | - Ye Jin Yang
- Department of Veterinary Physiology, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea; (M.J.K.); (Y.J.Y.); (J.W.H.); (J.-d.S.); (Y.Z.Y.)
| | - Ji Woong Heo
- Department of Veterinary Physiology, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea; (M.J.K.); (Y.J.Y.); (J.W.H.); (J.-d.S.); (Y.Z.Y.)
| | - Jae-dong Son
- Department of Veterinary Physiology, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea; (M.J.K.); (Y.J.Y.); (J.W.H.); (J.-d.S.); (Y.Z.Y.)
| | - Young Zoo You
- Department of Veterinary Physiology, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea; (M.J.K.); (Y.J.Y.); (J.W.H.); (J.-d.S.); (Y.Z.Y.)
| | - Ju-Hye Yang
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, 70 Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea;
| | - Kwang Il Park
- Department of Veterinary Physiology, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea; (M.J.K.); (Y.J.Y.); (J.W.H.); (J.-d.S.); (Y.Z.Y.)
| |
Collapse
|
13
|
Gowler PR, Arendt-Tranholm A, Turnbull J, Jha RR, Onion D, Kelly T, Kouraki A, Millns P, Gohir S, Franks S, Barrett DA, Valdes AM, Chapman V. Monocyte eukaryotic initiation factor 2 signaling differentiates 17-hydroxy-docosahexaenoic acid levels and pain. iScience 2025; 28:111862. [PMID: 39995860 PMCID: PMC11848799 DOI: 10.1016/j.isci.2025.111862] [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: 08/27/2024] [Revised: 11/04/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
Our goal was to probe the potential transcriptomic basis for the relationship between plasma levels of the specialized pro-resolving precursor, 17-hydroxy-docosahexaenoic acid (17-HDHA) and chronic pain. Participants with osteoarthritis (average age of 62.3, 60% were female, n = 30) were stratified by levels of 17-HDHA and self-reported pain scores. RNAs from CD14++/CD16-/CD66b-/HLA-DR+ (classical) monocytes were sequenced and differentially expressed mRNAs were identified with DESeq2. QIAGEN ingenuity pathway analysis identified the top ranked canonical biological pathway to be eukaryotic initiation factor 2 (EIF2) signaling (lower activation level in the low 17-HDHA-high pain group compared to the high 17-HDHA-low pain group (Z score -3)), followed by EIF4 and P70S6K signaling pathways and mTOR signaling. Our approach provides insight into the biological pathways contributing to the association between 17-HDHA and chronic osteoarthritis (OA) pain, identifying EIF2 signaling, with known roles in osteoclast differentiation, OA pathology, and pain, as a potential downstream target.
Collapse
Affiliation(s)
- Peter R.W. Gowler
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Asta Arendt-Tranholm
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - James Turnbull
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division and NIHR Nottingham Biomedical Research Centre, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Rakesh R. Jha
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division and NIHR Nottingham Biomedical Research Centre, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - David Onion
- Flow Cytometry Facility, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Tony Kelly
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Afroditi Kouraki
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Paul Millns
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Sameer Gohir
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Susan Franks
- School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - David A. Barrett
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division and NIHR Nottingham Biomedical Research Centre, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Ana M. Valdes
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Victoria Chapman
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
14
|
Ma X, Chin KY, Ekeuku SO. Anthocyanins and Anthocyanidins in the Management of Osteoarthritis: A Scoping Review of Current Evidence. Pharmaceuticals (Basel) 2025; 18:301. [PMID: 40143080 PMCID: PMC11944859 DOI: 10.3390/ph18030301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: The consumption of food rich in anthocyanins, a natural pigment found in plants, has been associated with improved joint health. However, systematic efforts to summarise the effects of anthocyanins and their deglycosylated forms, anthocyanidins, in managing osteoarthritis (OA) are lacking. This scoping review aims to comprehensively summarise the current evidence regarding the role of anthocyanins and anthocyanidins in OA management and highlights potential research areas. Methods: A comprehensive literature search was performed using PubMed, Scopus, and Web of Science in January 2025 to look for primary studies published in English, with the main objective of investigating the chondroprotective effects of anthocyanins and anthocyanidins, regardless of their study designs. Results: The seven included studies showed that anthocyanins and anthocyanidins suppressed the activation of inflammatory signalling, upregulated sirtuin-6 (cyanidin only), and autophagy (delphinidin only) in chondrocytes challenged with various stimuli (interleukin-1β, oxidative stress, or advanced glycation products). Anthocyanins also preserved cartilage integrity and increased the pain threshold in animal models of OA. No clinical trial was found in this field, suggesting a translation gap. Conclusions: In conclusion, anthocyanins and anthocyanidins are potential chondroprotective agents, but more investigations are required to overcome the gap in clinical translation.
Collapse
Affiliation(s)
- Xiaodong Ma
- Department of Traditional Chinese Medicine, Universiti Tunku Abdul Rahman, Kajang 43000, Malaysia;
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia
| | - Sophia Ogechi Ekeuku
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia
| |
Collapse
|
15
|
Dhaniya G, Mulay V, Kothari P, Sardar A, Chutani K, Parul, Verma S, Shukla S, Hingorani L, Trivedi R. Enrichment of the major bioavailable molecule glucuronated flavone TMMG in Spinacia oleracea ameliorates cartilage degeneration at a lower dose in ACLT-induced osteoarthritis. Food Funct 2025; 16:1469-1485. [PMID: 39898820 DOI: 10.1039/d4fo02128b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Spinacia oleracea extract (SOE) showed a protective effect on cartilage against osteoarthritis, with one of its compounds, 5,3',4'-trihydroxy-3-methoxy-6,7-methylenedioxy-flavone4'-glucuronide (TMMG), identified as a major bioavailable molecule with chondroprotective properties. Our recent study aimed to assess the potential of Spinacia oleracea enriched extract (SOEE) containing TMMG in alleviating osteoarthritis symptoms, facilitating easier determination of the human equivalent dose. Using an animal model simulating post-traumatic osteoarthritis, rats underwent anterior cruciate ligament transection (ACLT), with untreated animals serving as controls. Four weeks post-surgery, ACLT rats were randomly assigned for treatment with SOEE orally administered at doses of 10 and 20 mg kg-1 d-1 for four weeks. A positive control group was administered with crude SOE (125 mg kg-1 d-1; ∼1% TMMG). Two days prior to the termination of the animal study, behavioural analysis was done through open field activity and rotarod tests to assess the locomotive activity. Furthermore, data analysis was done through HPLC (high-performance liquid chromatography). Additional investigations corroborated chondroprotective effects via gross morphology of the knee joint, histological assessment of tibial articular cartilage, serum biochemical analysis of cartilage degradation markers, and micro-CT (micro-computed tomography). In conclusion, SOEE at 10 mg kg-1 d-1 demonstrated superior chondroprotective efficacy when compared to its 20 mg kg-1 d-1 dosage as well as SOE alone. Further investigation could lead to establishing a human equivalent dose of 1.522 mg kg-1 for osteoarthritis treatment.
Collapse
Affiliation(s)
- Geeta Dhaniya
- Endocrinology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
| | - Vallabh Mulay
- Pharmanza Herbal Pvt Ltd, Anand, Gujarat, 388435, India
| | - Priyanka Kothari
- Endocrinology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, India
| | - Anirban Sardar
- Endocrinology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kunal Chutani
- Endocrinology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Parul
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Shikha Verma
- Endocrinology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shubha Shukla
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Lal Hingorani
- Pharmanza Herbal Pvt Ltd, Anand, Gujarat, 388435, India
| | - Ritu Trivedi
- Endocrinology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
16
|
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
|
17
|
Xiong L. SUMOylated GLUT1 inhibited the glycometabolism disorder in chondroctyes during osteoarthritis. Glycoconj J 2025; 42:41-52. [PMID: 39797965 DOI: 10.1007/s10719-024-10176-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025]
Abstract
Reduction of glucose transporter 1 (GLUT1), even deletion, may results in cartilage fibrosis and osteoarthritis. This study aims to investigate the SUMOylation of GLUT1 in osteoarthritis through small ubiquitin-like modifier 1(SUMO1), and explore the role of SUMOylated GLUT1 in glycometabolism, proliferation and apoptosis in chondrocytes. Human chondrocytes were incubated with 10 ng/mL of IL-1β to mimic osteoarthritis in vitro. GLUT1, SUMO1 and Chondrocyte-related genes including COL2A1, MMP13 and ADAMTS4 were evaluated using western blot. Cell viability and cell apoptosis of chondrocytes were measured by cell counting kit-8 assay and flow cytometry, respectively. The changes in glycometabolism were evaluated using extracellular acidification rate (ECAR) and glucose uptake assay. Co-immunoprecipitation (Co-IP) was used to verify the interaction between GLUT1 and SUMO1. The stabilization role of SUMO1 in GLUT1 was determined by cycloheximide assay. IL-1β induced the decrease of GLUT1, cell viability, ECAR, glucose uptake and COL2A1 and the increase of cell apoptosis, MMP13 and ADAMTS4 in chondrocytes. However, overexpression of SUMO1 led to the reduction of cell apoptosis, MMP13 and ADAMTS4 and the elevation of GLUT1, cell viability, ECAR, glucose uptake and COL2A1 in IL-1β-stimulated chondrocytes. There was SUMOylation sites on GLUT1. Intriguingly, SUMO1 was significantly enriched in GLUT1 using Co-IP assay, and stabilized GLUT1 in chondrocytes. SUMO1-mediated SUMOylation is capable of stabilizing GLUT1 to inhibit glycometabilsm disorder and cell apoptosis in IL-1β-stimulated chondrocytes.
Collapse
Affiliation(s)
- Liwei Xiong
- Department of Orthopaedics, Nanchang People's Hospital (The Third Hospital of Nanchang), Nanchang City, Jiangxi Province, China.
- Nanchang People's Hospital (The Third Hospital of Nanchang), No. 2, Xiangshan South Road, Nanchang City, Jiangxi Province, 330003, China.
| |
Collapse
|
18
|
Dulic O, Abazovic D, Matijevic S, Rasovic P, Obradovic M, Bjelobrk M, Tosic M, Lalic I, Baljak B, Milinkov M. Quality of life changes in patients suffering from knee osteoarthritis treated with bone marrow aspirate concentrate, platelet-rich plasma and hyaluronic acid injections. Regen Med 2025; 20:87-96. [PMID: 40028743 PMCID: PMC11951690 DOI: 10.1080/17460751.2025.2472589] [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: 09/19/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND This study aimed to compare the effects of different treatments on quality of life in knee osteoarthritis patients. It focused on three therapies: bone marrow aspirate concentrate (BMAC), platelet-rich plasma (PRP), and hyaluronic acid (HA). METHODOLOGY The trial was conducted at a single center with 175 patients over a 12-month period with the knee OA, KL grade II-IV. Outcomes were measured using the WOMAC (Western Ontario and McMaster Universities Osteoarthritis Index) and SF-36 scales, which assess physical and emotional well-being. Linear mixed models (LMMs) were used to analyze which treatment had the most positive impact on quality of life. RESULTS Patients treated with BMAC showed the most substantial improvement, particularly in physical health and mobility (p ≤ 0,001). PRP outperformed HA in some aspects, but BMAC consistently led to greater gains. The most notable enhancements were seen in areas like role limitations due to physical health and overall physical functioning. CONCLUSIONS The study suggested that BMAC treatment may contribute to improved quality of life in patients with knee osteoarthritis, particularly in terms of physical function. The correlation between WOMAC and SF-36 scores supports these findings, indicating a potential role for BMAC in enhancing mobility. CLINICAL TRIAL REGISTRATION NCT03825133 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Oliver Dulic
- Medical Faculty, University of Novi Sad, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | | | | | - Predrag Rasovic
- Medical Faculty, University of Novi Sad, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Mirko Obradovic
- Medical Faculty, University of Novi Sad, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Mile Bjelobrk
- Medical Faculty, University of Novi Sad, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Milan Tosic
- Medical Faculty, University of Novi Sad, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Ivica Lalic
- Faculty of Farmacy, University Business Academy, Novi Sad, Serbia
| | - Branko Baljak
- Medical Faculty, University of Novi Sad, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Milan Milinkov
- Medical Faculty, University of Novi Sad, University Clinical Center of Vojvodina, Novi Sad, Serbia
| |
Collapse
|
19
|
Kaneta H, Shoji T, Kato Y, Shozen H, Ueki S, Morita H, Kozuma Y, Adachi N. Relationship Between the Subchondral Trabecular Bone Microstructure in the Hip Joint and Pain in Patients with Hip Osteoarthritis. Cartilage 2024:19476035241302978. [PMID: 39651681 PMCID: PMC11626549 DOI: 10.1177/19476035241302978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/15/2024] [Accepted: 11/10/2024] [Indexed: 12/11/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the relationship between clinical findings and the trabecular microstructure of the subchondral bone in patients with hip osteoarthritis (OA) due to developmental dysplasia of the hip (DDH) using multidetector row computed tomography (MDCT). DESIGN A total of 63 patients (69 hips) with OA due to DDH were retrospectively reviewed, with 12 healthy controls being included for comparison. Clinical evaluation was performed using the Japanese Orthopaedic Association Hip Disease Evaluation Questionnaire (JHEQ). The trabecular bone microstructure was analyzed using MDCT. Regions of interest in the subchondral trabecular bones of the acetabulum and femoral head were defined in the coronal view, and various trabecular microstructural parameters were evaluated. RESULTS Bone volume fraction (BV/TV) and trabecular thickness (Tb.Th) exhibited a significant positive correlation with the OA stage, whereas trabecular separation (Tb.Sp) showed a negative correlation. In addition, BV/TV and Tb.Th were negatively correlated with the JHEQ total and pain scores, whereas Tb.Sp was positively correlated with the pain score in all regions. CONCLUSIONS This is the first study to evaluate the bone microstructure and its relationship with clinical findings in patients with hip OA due to DDH. Our findings suggest that as OA progresses, osteosclerotic changes increase in the acetabulum and femoral head; these changes are associated with worsening clinical symptoms, particularly pain. Targeting the subchondral bone may emerge as a novel treatment strategy for patients with OA due to DDH; nevertheless, further comprehensive studies are required.
Collapse
Affiliation(s)
- Hiroki Kaneta
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takeshi Shoji
- Department of Artificial Joints and Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuichi Kato
- Department of Orthopaedic Surgery, Chugoku Rosai Hospital, Kure, Japan
| | - Hideki Shozen
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinichi Ueki
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroyuki Morita
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yosuke Kozuma
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
20
|
Chen Y, Liang R, Zheng X, Fang D, Lu WW, Chen Y. Identification of ZNF652 as a Diagnostic and Therapeutic Target in Osteoarthritis Using Machine Learning. J Inflamm Res 2024; 17:10141-10161. [PMID: 39649418 PMCID: PMC11624598 DOI: 10.2147/jir.s488841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/17/2024] [Indexed: 12/10/2024] Open
Abstract
Purpose Osteoarthritis (OA) is the most common degenerative joint disease. However, its etiology remains largely unknown. Zinc Finger Protein 652 (ZNF652) is a transcription factor implicated in various biological processes. Nevertheless, its role in OA has not been elucidated. Methods The search term "osteoarthritis" was utilized to procure transcriptome data relating to OA patients and healthy people from the Gene Expression Omnibus (GEO) database. Then a screening process was initiated to identify differentially expressed genes (DEGs). The DEGs were discerned using three distinct machine learning methods. The accuracy of these DEGs in diagnosing OA was evaluated using the Receiver Operating Characteristic (ROC) Curve. A competitive endogenous RNA (ceRNA) visualization network was established to delve into potential regulatory targets. The ZNF652 expression was confirmed in the cartilage of OA rats using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting (WB) and analyzed using an independent t-test. Results ZNF652 was identified as a DEG and exhibited the highest diagnostic value for OA according to the ROC analysis. The GO and KEGG enrichment analyses suggest that ZNF652 plays a vital role in OA development through processes including nitric oxide anabolism, macrophage proliferation, immune response, and the PI3K/Akt and the MAPK signaling pathways. The increased expression of ZNF652 in OA was validated in qRT-PCR (1.193 ± 0.005 vs 1.000 ± 0.005, p < 0.001) and WB (0.981 ± 0.055 vs 0.856 ± 0.026, p = 0.012) analysis. Conclusion ZNF652 was found to be related to OA pathogenesis and can potentially serve as a diagnostic and therapeutic target of OA. The underlying mechanism is that ZNF652 was related to nitric oxide anabolism, macrophage proliferation, various signaling pathways, and immune cells and their functions in OA. Nevertheless, the findings need to be confirmed in clinical trials and the molecular mechanism requires further study.
Collapse
Affiliation(s)
- Yeping Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Rongyuan Liang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Xifan Zheng
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Dalang Fang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Youjiang Medical College of Nationalities, Baise, Guangxi, People’s Republic of China
| | - William W Lu
- Department of Orthopedics and Traumatology, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Yan Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| |
Collapse
|
21
|
Henriques J, Berenbaum F, Mobasheri A. Obesity-induced fibrosis in osteoarthritis: Pathogenesis, consequences and novel therapeutic opportunities. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100511. [PMID: 39483440 PMCID: PMC11525450 DOI: 10.1016/j.ocarto.2024.100511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/26/2024] [Accepted: 08/12/2024] [Indexed: 11/03/2024] Open
Abstract
Osteoarthritis (OA) is a significant global burden, affecting more than half a billion people across the world. It is characterized by degeneration and loss of articular cartilage, synovial inflammation, and subchondral bone sclerosis, leading to pain and functional impairment. After age, obesity is a major modifiable risk factor for OA, and it has recently been identified as a chronic disease by the World Health Organization (WHO). Obesity is associated with high morbidity and mortality, imposing a significant cost on individuals and society. Obesity increases the risk of knee OA through increased joint loading, altered body composition, and elevated pro-inflammatory adipokines in the systemic circulation. Moreover, obesity triggers fibrotic processes in different organs and tissues, including those involved in OA. Fibrosis in OA refers to the abnormal accumulation of fibrous tissue within and around the joints. It can be driven by increased adiposity, low-grade inflammation, oxidative stress, and metabolic alterations. However, the clinical outcomes of fibrosis in OA are unclear. This review focuses on the link between obesity and OA, explores the mechanism of obesity-driven fibrosis, and examines potential therapeutic opportunities for targeting fibrotic processes in OA.
Collapse
Affiliation(s)
- João Henriques
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Francis Berenbaum
- Sorbonne University, Paris, France
- Department of Rheumatology, Saint-Antoine Hospital, Assistance Publique-Hopitaux de Paris, Paris, France
- INSERM CRSA, Paris, France
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| |
Collapse
|
22
|
Xu W, Zhang Y, Li L, Pan L, Lu L, Zhi S, Li W. Osteocyte-derived exosomes regulate the DLX2/wnt pathway to alleviate osteoarthritis by mediating cartilage repair. Autoimmunity 2024; 57:2364686. [PMID: 38946534 DOI: 10.1080/08916934.2024.2364686] [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: 09/19/2023] [Accepted: 06/02/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Chondrocyte viability, apoptosis, and migration are closely related to cartilage injury in osteoarthritis (OA) joints. Exosomes are identified as potential therapeutic agents for OA. OBJECTIVE This study aimed to investigate the role of exosomes derived from osteocytes in OA, particularly focusing on their effects on cartilage repair and molecular mechanisms. METHODS An injury cell model was established by treating chondrocytes with IL-1β. Cartilage repair was evaluated using cell counting kit-8, flow cytometry, scratch test, and Western Blot. Molecular mechanisms were analyzed using quantitative real-time PCR, bioinformatic analysis, and Western Blot. An OA mouse model was established to explore the role of exosomal DLX2 in vivo. RESULTS Osteocyte-released exosomes promoted cell viability and migration, and inhibited apoptosis and extracellular matrix (ECM) deposition. Moreover, exosomes upregulated DLX2 expression, and knockdown of DLX2 activated the Wnt pathway. Additionally, exosomes attenuated OA in mice by transmitting DLX2. CONCLUSION Osteocyte-derived exosomal DLX2 alleviated IL-1β-induced cartilage repair and inactivated the Wnt pathway, thereby alleviating OA progression. The findings suggested that osteocyte-derived exosomes may hold promise as a treatment for OA.
Collapse
Affiliation(s)
- Wenjuan Xu
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Yuanyuan Zhang
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Lijuan Li
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Liyan Pan
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Li Lu
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Shenshen Zhi
- Department of Blood Transfusion, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Wei Li
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| |
Collapse
|
23
|
Jones R, Gilbert SJ, Christofides SR, Mason DJ. Osteocytes contribute to sex-specific differences in osteoarthritic pain. Front Endocrinol (Lausanne) 2024; 15:1480274. [PMID: 39574959 PMCID: PMC11579924 DOI: 10.3389/fendo.2024.1480274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024] Open
Abstract
Osteoarthritic (OA) pain affects 18% of females and 9.6% of males aged over 60 worldwide, with 62% of all OA patients being women. The molecular drivers of sex-based differences in OA are unknown. Bone is intricately coupled with the sensory nervous system and one of the only joint tissues known to show changes that correlate with patient pain in OA. There are fundamental sex differences in pain sensation and bone biology which may be intrinsic to OA disease progression, however these differences are vastly under researched. We have utilised three data sets to investigate the hypothesis that potential mediators responsible for sex dependent pain mechanisms displayed in OA are derived from mechanically stimulated osteocytes. Our published dataset of the in vitro human osteocyte mechanosome was independently compared with published data from, sex-based gene expression differences in human long bone, the sex-based gene expression differences during the skeletal maturation of the mouse osteocyte transcriptome and sex specific OA risk factors and effector genes in a large human GWAS. 80 of the 377 sex-specific genes identified in the mouse osteocyte transcriptome were mechanically regulated in osteocytes with enrichment associated with neural crest migration and axon extension, and DISEASES analysis enrichment for the rheumatoid arthritis pathway. 3861 mechanically regulated osteocytic genes displayed sex-specific differences in human long bone with enrichment for genes associated with the synapse, sensory perception of pain, axon guidance, immune responses, distal peripheral sensory neuropathy, sensory neuropathy, and poor wound healing. 32 of 77 effector genes and 1 of 3 female specific OA risk factor genes identified in the human GWAS were differentially expressed in the osteocyte mechanosome and male and female bone. This analysis lends support to the hypothesis that mechanically regulated genes in osteocytes could influence sex specific differences in osteoarthritic pain and highlights pain pathways with approved drugs that could potentially treat elevated pain susceptibility in females with OA.
Collapse
Affiliation(s)
| | | | | | - Deborah J. Mason
- Biomechanics and Bioengineering Research Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
24
|
Liao J, Gu Q, Liu Z, Wang H, Yang X, Yan R, Zhang X, Song S, Wen L, Wang Y. Edge advances in nanodrug therapies for osteoarthritis treatment. Front Pharmacol 2024; 15:1402825. [PMID: 39539625 PMCID: PMC11559267 DOI: 10.3389/fphar.2024.1402825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
As global population and lifestyles change, osteoarthritis (OA) is becoming a major healthcare challenge world. OA, a chronic condition characterized by inflammatory and degeneration, often present with joint pain and can lead to irreversible disability. While there is currently no cure for OA, it is commonly managed using nonsteroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, and glucosamine. Although these treatments can alleviate symptoms, it is difficult to effectively deliver and sustain therapeutic agents within joints. The emergence of nanotechnology, particularly in form of smart nanomedicine, has introduced innovative therapeutic approaches for OA treatment. Nanotherapeutic strategies offer promising advantages, including more precise targeting of affected areas, prolonged therapeutic effects, enhanced bioavailability, and reduced systemic toxicity compared to traditional treatments. While nanoparticles show potential as a viable delivery system for OA therapies based on encouraging lab-based and clinical trials results, there remails a considerable gap between current research and clinical application. This review highlights recent advances in nanotherapy for OA and explore future pathways to refine and optimize OA treatments strategies.
Collapse
Affiliation(s)
- Jinfeng Liao
- Department of Dermatology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Qingjia Gu
- Department of ENT, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Zheng Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Xian Yang
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongkai Yan
- Department of Radiology, Ohio state university, Columbus, OH, United States
| | - Xiaofeng Zhang
- Greenwich Hospital, Yale New Haven Health, Greenwich, CT, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Lebin Wen
- Department of Thyroid, Sichuan Second Hospital of TCM, Chengdu, China
| | - Yi Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
25
|
Maouche A, Boumediene K, Baugé C. Bioactive Compounds in Osteoarthritis: Molecular Mechanisms and Therapeutic Roles. Int J Mol Sci 2024; 25:11656. [PMID: 39519204 PMCID: PMC11546619 DOI: 10.3390/ijms252111656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA) is the most common and debilitating form of arthritis. Current therapies focus on pain relief and efforts to slow disease progression through a combination of drug and non-drug treatments. Bioactive compounds derived from plants show significant promise due to their anti-inflammatory, antioxidant, and tissue-protective properties. These natural compounds can help regulate the inflammatory processes and metabolic pathways involved in OA, thereby alleviating symptoms and potentially slowing disease progression. Investigating the efficacy of these natural agents in treating osteoarthritis addresses a growing demand for natural health solutions and creates new opportunities for managing this increasingly prevalent age-related condition. The aim of this review is to provide an overview of the use of some bioactive compounds from plants in modulating the progression of osteoarthritis and alleviating associated pain.
Collapse
Affiliation(s)
| | | | - Catherine Baugé
- UR7451 BIOCONNECT, Université de Caen Normandie, 14032 Caen, France; (A.M.); (K.B.)
| |
Collapse
|
26
|
Han J, Zhang J, Zhang X, Luo W, Liu L, Zhu Y, Liu Q, Zhang XA. Emerging role and function of Hippo-YAP/TAZ signaling pathway in musculoskeletal disorders. Stem Cell Res Ther 2024; 15:386. [PMID: 39468616 PMCID: PMC11520482 DOI: 10.1186/s13287-024-04011-9] [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: 08/06/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
Hippo pathway is an evolutionarily conservative key pathway that regulates organ size and tissue regeneration by regulating cell proliferation, differentiation and apoptosis. Yes-associated protein 1 (YAP)/ WW domain-containing transcription regulator 1 (TAZ) serves as a pivotal transcription factor within the Hippo signaling pathway, which undergoes negative regulation by the Hippo pathway. The expression of YAP/TAZ affects various biological processes, including differentiation of osteoblasts (OB) and osteoclasts (OC), cartilage homeostasis, skeletal muscle development, regeneration and quality maintenance. At the same time, the dysregulation of the Hippo pathway can concurrently contribute to the development of various musculoskeletal disorders, including bone tumors, osteoporosis (OP), osteoarthritis (OA), intervertebral disc degeneration (IDD), muscular dystrophy, and rhabdomyosarcoma (RMS). Therefore, targeting the Hippo pathway has emerged as a promising therapeutic strategy for the treatment of musculoskeletal disorders. The focus of this review is to elucidate the mechanisms by which the Hippo pathway maintains homeostasis in bone, cartilage, and skeletal muscle, while also providing a comprehensive summary of the pivotal role played by core components of this pathway in musculoskeletal diseases. The efficacy and feasibility of Hippo pathway-related drugs for targeted therapy of musculoskeletal diseases are also discussed in our study. These endeavors offer novel insights into the application of Hippo signaling in musculoskeletal disorders, providing effective therapeutic targets and potential drug candidates for treating such conditions.
Collapse
Affiliation(s)
- Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Jiale Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Xiaoyi Zhang
- College of Second Clinical Medical, China Medical University, Shenyang, 110122, China
| | - Wenxin Luo
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Lifei Liu
- Department of Rehabilitation, The People's Hospital of Liaoning Province, Shenyang, 110016, China
| | - Yuqing Zhu
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Qingfeng Liu
- Department of General Surgery, Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China.
| |
Collapse
|
27
|
Liang J, Hu J, Hong X, Zhou M, Xia G, Hu L, Luo S, Quan K, Yan J, Wang S, Fan S. Amentoflavone maintaining extracellular matrix homeostasis and inhibiting subchondral bone loss in osteoarthritis by inhibiting ERK, JNK and NF-κB signaling pathways. J Orthop Surg Res 2024; 19:662. [PMID: 39407273 PMCID: PMC11481797 DOI: 10.1186/s13018-024-05075-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Amentoflavone (AF), a plant biflavone isolated from Selaginella sinensis ethanol extract, is characterized by anti-inflammatory and anti-oxidant properties. According to previous studies, inflammation and oxidative stress are closely related to the pathophysiology of osteoarthritis (OA). However, the effects and mechanisms of AF on OA have not been elucidated.To investigate the inhibitory effects and its molecular mechanism of AF on extracellular matrix (ECM) degradation stimulated by IL-1β as well as subchondral bone loss induced by RANKL in mice chondrocytes. Quantitative PCR was used to detect the mRNA expression of genes related to inflammation, ECM, and osteoclast differentiation. Protein expression level of iNOS, COX-2, MMP13, ADAMTS5, COL2A1, SOX9, NFATc1, c-fos, JNK, ERK, P65, IκBα was measured by western blotting. The levels of TNF-α and IL-6 in the supernatants were measured by ELISA. The amount of ECM in chondrocytes was measured using toluidine blue staining. The levels of Aggrecan and Col2a1 in chondrocytes were measured using immunofluorescence. Tartrate-resistant acid phosphatase (TRAP) staining, F-actin staining and immunofluorescence were used to detect the effect of AF on osteoclast differentiation and bone resorption. The effect of AF on destabilization of the medial meniscus (DMM)-induced OA mice can be detected in hematoxylin-eosin (H&E) staining, Safranin O green staining and immunohistochemistry.AF might drastically attenuated IL-1β-stimulated inflammation and reduction of ECM formation by blocking ERK and NF-κB signaling pathways in chondrocytes. Meanwhile, AF suppressed the formation of osteoclasts and the resorption of bone function induced by RANKL. In vivo, AF played a protective role by stabilizing cartilage ECM and inhibiting subchondral bone loss in destabilization of the medial meniscus (DMM)-induced OA mice, further proving its protective effect in the development of OA. Our study show that AF alleviated OA by suppressing ERK, JNK and NF-κB signaling pathways in OA models in vitro and DMM-induced OA mice, suggesting that AF might be a potential therapeutic agent in the treatment of OA.
Collapse
Affiliation(s)
- Jianhui Liang
- Department of Orthopedics, Nanchang Hongdu Hospital of TCM, Nanchang, China
| | - Jiawei Hu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xin Hong
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Ming Zhou
- Department of Orthopedics, Nanchang Hongdu Hospital of TCM, Nanchang, China
| | - Guoming Xia
- Department of Orthopedics, Nanchang Hongdu Hospital of TCM, Nanchang, China
| | - Liangshen Hu
- Department of Orthopedics, Nanchang Hongdu Hospital of TCM, Nanchang, China
| | - Song Luo
- Department of Orthopedics, Nanchang Hongdu Hospital of TCM, Nanchang, China
| | - Kun Quan
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Jianbin Yan
- Department of Orthopedics, Nanchang Hongdu Hospital of TCM, Nanchang, China
| | - Song Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China.
| | - Shaoyong Fan
- Department of Orthopedics, Nanchang Hongdu Hospital of TCM, Nanchang, China.
| |
Collapse
|
28
|
Yu X, Zhuang R, Jin P. Evaluation of the efficacy after Total Knee Arthroplasty by Gait analysis in patients with Knee Osteoarthritis: a meta-analysis. J Orthop Surg Res 2024; 19:612. [PMID: 39343975 PMCID: PMC11441000 DOI: 10.1186/s13018-024-05091-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Total knee replacement (TKA) is a frequent modality performed in patients with knee osteoarthritis (OA). The aim of this study was to perform a meta-analysis and systematic review to evaluate the efficacy after TKA by gait analysis in patients with OA. METHODS PubMed, EMBASE, the Cochrane library, and Web of Science were searched for relevant studies from inception to July 2024. STATA SE 14.0 software was used for statistical analysis according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA 2020) guideline. RESULTS A total of 2525 reports were identified with 24 studies meeting pre-designed inclusion criteria. Several gait parameters were investigated. In patients with knee OA after TKA, there existed an increase in the Max knee flexion (WMD, 3.12; 95% CI, 0.93 to 5.32; I2 = 73.9%, P < 0.001), the Cadence (WMD, 4.05; 95% CI, 2.28 to 5.82; I2 = 48.9%, P = 0.068), the stride length (WMD, 0.05; 95% CI, 0.01 to 0.09; I2 = 77.1%, P < 0.001), the walking speed (WMD, 0.08; 95% CI, 0.02 to 0.14; I2 = 93.3%, P < 0.001), and the step length (WMD, 0.04; 95% CI, 0.00 to 0.07; I2 = 89.3%, P < 0.001) while a decrease in the double support time (WMD, -0.04; 95% CI, - 0.08 to -0.01; I2 = 0.0%, P = 0.585). Besides, no statistically significant differences were observed in the Knee range of motion (ROM), the Max knee rotation at stance phase, the Max knee extension, the step width, the stride time and the step time. Sensitivity analysis showed that all the results were robust. CONCLUSIONS In summary, the study found that, in patients with knee OA undergoing TKA may have great effects on improving gait parameters. If there are more high-quality studies in the future, we should make a more comprehensive evaluation of walking function by gait analysis together with other evaluation systems such as muscle strength and proprioception measurement.
Collapse
Affiliation(s)
- Xinfeng Yu
- Department of Orthopedics, Sanmen People's Hospital, Taizhou, Zhejiang, 317100, China
| | - Rujie Zhuang
- Department of Orthopedics, Quzhou TCM Hospital at the Junction of Four Provinces Affiliated to Zhejiang Chinese Medical University, Quzhou, Zhejiang, 324000, China.
| | - Peng Jin
- Department of Orthopedics, Sanmen People's Hospital, Taizhou, Zhejiang, 317100, China
| |
Collapse
|
29
|
Georgescu B, Cristea AE, Oprea D, Lupu AA, Stanciu LE, Borgazi E, Caraban BM, Ciortea VM, Irsay L, Iliescu MG. Current Evidence on and Clinical Implications of Vitamin D Levels in Pain and Functional Management of Knee Osteoarthritis: A Systematic Review. Clin Pract 2024; 14:1997-2012. [PMID: 39451873 PMCID: PMC11506653 DOI: 10.3390/clinpract14050158] [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: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Osteoarthritis is a common chronic disease that affects quality of life and increases public health costs. Knee osteoarthritis is a frequent form, marked by joint degeneration, pain, stiffness, and functional restrictions. Factors such as age, genetics, joint injuries, obesity, and vitamin D deficiency can affect knee osteoarthritis progression. While the exact link between vitamin D and osteoarthritis is still being studied, recent research indicates that low vitamin D levels might influence the articular cartilage's structure and function, potentially accelerating osteoarthritis. This review aims to analyze the last decade of research on vitamin D's role in osteoarthritis. METHODS A systematic review of the literature was conducted in accordance with the PRISMA guidelines (Preferred Reporting Items for Systematic Reviews and Meta-Analyses). Relevant studies from the last ten years were included to evaluate the association between vitamin D levels and knee osteoarthritis. The inclusion criteria were studies examining the role of vitamin D in cartilage health and osteoarthritis progression and the potential clinical implications for disease management. RESULTS This review identified a variety of studies exploring the connection between vitamin D and osteoarthritis, with mixed findings. CONCLUSIONS The relationship between vitamin D and knee osteoarthritis remains inconclusive, highlighting the need for further research. An updated evaluation of the literature is crucial for osteoarthritis management strategies and to potentially include vitamin D supplementation in therapeutic protocols.
Collapse
Affiliation(s)
- Bianca Georgescu
- Medical Doctoral School, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania; (B.G.); (A.E.C.)
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania; (D.O.); (L.-E.S.)
| | - Adelina Elena Cristea
- Medical Doctoral School, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania; (B.G.); (A.E.C.)
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania; (D.O.); (L.-E.S.)
| | - Doinița Oprea
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania; (D.O.); (L.-E.S.)
| | - Andreea Alexandra Lupu
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania; (D.O.); (L.-E.S.)
| | - Liliana-Elena Stanciu
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania; (D.O.); (L.-E.S.)
| | - Erdin Borgazi
- Department of Orthopedics, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania;
| | - Bogdan Marian Caraban
- Department of Plastic Surgery, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania;
| | - Viorela Mihaela Ciortea
- Department of Rehabilitation Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (V.M.C.); (L.I.)
| | - Laszlo Irsay
- Department of Rehabilitation Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (V.M.C.); (L.I.)
| | - Mădălina Gabriela Iliescu
- Medical Doctoral School, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania; (B.G.); (A.E.C.)
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Ovidius University of Constanta, 1 University Alley, Campus—Corp B, 900470 Constanta, Romania; (D.O.); (L.-E.S.)
| |
Collapse
|
30
|
Matsukura K, Kondo M, Metzler NF, Ford AJ, Maak TG, Hutchinson DT, Wang AA, Sato M, Grainger DW, Okano T. Regenerative Variability of Human Juvenile Chondrocyte Sheets From Different Cell Donors in an Athymic Rat Knee Chondral Defect Model. Cartilage 2024:19476035241277946. [PMID: 39319855 PMCID: PMC11556591 DOI: 10.1177/19476035241277946] [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] [Indexed: 09/26/2024] Open
Abstract
PURPOSE This study aimed to establish a combined histological assessment system of neo-cartilage outcomes and to evaluate variations in an established rat defect model treated with human juvenile cartilage-derived chondrocyte (JCC) sheets fabricated from various donors. METHODS JCCs were isolated from the polydactylous digits of eight patients. Passage 2 (P2) JCC sheets from all donors were transplanted into nude rat chondral defects for 4 weeks (27 nude rats in total). Defect-only group served as control. Histological samples were stained for safranin O, collagen 1 (COL1), and collagen 2 (COL2). (1) All samples were scored, and correlation coefficients for each score were calculated. (2) Donors were divided into "more effective" and "less effective" groups based on these scores. Then, differences between each group in each category of modified O'Driscoll scoring were evaluated. RESULTS (1) Modified O'Driscoll scores were negatively correlated with %COL1 area, and positively correlated with %COL2 area and COL2/1 ratio. (2) Four of 8 donors exhibited significantly higher modified O'Driscoll scores and %COL2 areas. JCC donors were divided into two groups by average score values. Significant differences between the two groups were observed in modified O'Driscoll categories of "Nature of predominant tissue," "Reconstruction of subchondral bone," and "Safranin O staining." CONCLUSION The combined histological evaluation method is useful for detailed in vivo efficacy assessments of cartilage defect regeneration models. Variations in histological scores among juvenile cartilage-derived chondrocyte donors were correlated to the quality of regenerated cartilage hyaline structure and subchondral bone remodeling observed in the nude rat defect model.
Collapse
Affiliation(s)
- Keisuke Matsukura
- Cell Sheet Tissue Engineering Center, Department of Molecular Pharmaceutics, Health Sciences, The University of Utah, Utah, USA
- Department of Orthopedic, Asahikawa Medical University, Asahikawa, Japan
| | - Makoto Kondo
- Cell Sheet Tissue Engineering Center, Department of Molecular Pharmaceutics, Health Sciences, The University of Utah, Utah, USA
| | - Nicolas F. Metzler
- Cell Sheet Tissue Engineering Center, Department of Molecular Pharmaceutics, Health Sciences, The University of Utah, Utah, USA
- Department of Biomedical Engineering, The University of Utah, Salt Lake City, UT, USA
| | - Adam J. Ford
- Cell Sheet Tissue Engineering Center, Department of Molecular Pharmaceutics, Health Sciences, The University of Utah, Utah, USA
| | - Travis G. Maak
- Department of Orthopaedics, School of Medicine, The University of Utah, Salt Lake City, UT, USA
| | - Douglas T. Hutchinson
- Department of Orthopaedics, School of Medicine, The University of Utah, Salt Lake City, UT, USA
- Pediatric Orthopaedic Surgery, Intermountain Primary Children’s Hospital, Salt Lake City, UT, USA
| | - Angela A. Wang
- Department of Orthopaedics, School of Medicine, The University of Utah, Salt Lake City, UT, USA
- Pediatric Orthopaedic Surgery, Intermountain Primary Children’s Hospital, Salt Lake City, UT, USA
| | - Masato Sato
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - David W. Grainger
- Cell Sheet Tissue Engineering Center, Department of Molecular Pharmaceutics, Health Sciences, The University of Utah, Utah, USA
- Department of Biomedical Engineering, The University of Utah, Salt Lake City, UT, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center, Department of Molecular Pharmaceutics, Health Sciences, The University of Utah, Utah, USA
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Shinjuku-ku, Japan
| |
Collapse
|
31
|
Durrani IA, John P, Bhatti A, Khan JS. Network medicine based approach for identifying the type 2 diabetes, osteoarthritis and triple negative breast cancer interactome: Finding the hub of hub genes. Heliyon 2024; 10:e36650. [PMID: 39281650 PMCID: PMC11401126 DOI: 10.1016/j.heliyon.2024.e36650] [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: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/18/2024] Open
Abstract
The increasing prevalence of multi-morbidities, particularly the incidence of breast cancer in diabetic/osteoarthritic patients emphasize on the need for exploring the underlying molecular mechanisms resulting in carcinogenesis. To address this, present study employed a systems biology approach to identify switch genes pivotal to the crosstalk between diseased states resulting in multi-morbid conditions. Hub genes previously reported for type 2 diabetes mellitus (T2DM), osteoarthritis (OA), and triple negative breast cancer (TNBC), were extracted from published literature and fed into an integrated bioinformatics analyses pipeline. Thirty-one hub genes common to all three diseases were identified. Functional enrichment analyses showed these were mainly enriched for immune and metabolism associated terms including advanced glycation end products (AGE) pathways, cancer pathways, particularly breast neoplasm, immune system signalling and adipose tissue. The T2DM-OA-TNBC interactome was subjected to protein-protein interaction network analyses to identify meta hub/clustered genes. These were prioritized and wired into a three disease signalling map presenting the enriched molecular crosstalk on T2DM-OA-TNBC axes to gain insight into the molecular mechanisms underlying disease-disease interactions. Deciphering the molecular bases for the intertwined metabolic and immune states may potentiate the discovery of biomarkers critical for identifying and targeting the immuno-metabolic origin of disease.
Collapse
Affiliation(s)
- Ilhaam Ayaz Durrani
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, 44000, Pakistan
| | - Peter John
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, 44000, Pakistan
| | - Attya Bhatti
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, 44000, Pakistan
| | | |
Collapse
|
32
|
Zhu X, Cao M, Li K, Chan YT, Chan HF, Mak YW, Yao H, Sun J, Ong MTY, Ho KKW, Lee CW, Lee OKS, Yung PSH, Jiang Y. Intra-articular sustained-release of pirfenidone as a disease-modifying treatment for early osteoarthritis. Bioact Mater 2024; 39:255-272. [PMID: 38832304 PMCID: PMC11145079 DOI: 10.1016/j.bioactmat.2024.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/28/2024] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Osteoarthritis (OA) is a major clinical challenge, and effective disease-modifying drugs for OA are still lacking due to the complicated pathology and scattered treatment targets. Effective early treatments are urgently needed to prevent OA progression. The excessive amount of transforming growth factor β (TGFβ) is one of the major causes of synovial fibrosis and subchondral bone sclerosis, and such pathogenic changes in early OA precede cartilage damage. Herein we report a novel strategy of intra-articular sustained-release of pirfenidone (PFD), a clinically-approved TGFβ inhibitor, to achieve disease-modifying effects on early OA joints. We found that PFD effectively restored the mineralization in the presence of excessive amount of TGFβ1 (as those levels found in patients' synovial fluid). A monthly injection strategy was then designed of using poly lactic-co-glycolic acid (PLGA) microparticles and hyaluronic acid (HA) solution to enable a sustained release of PFD (the "PLGA-PFD + HA" strategy). This strategy effectively regulated OA progression in destabilization of the medial meniscus (DMM)- induced OA mice model, including preventing subchondral bone loss in early OA and subchondral bone sclerosis in late OA, and reduced synovitis and pain with cartilage preservation effects. This finding suggests the promising clinical application of PFD as a novel disease-modifying OA drug.
Collapse
Affiliation(s)
- Xiaobo Zhu
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China
- Department of Orthopaedic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, China
| | - Mingde Cao
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China
| | - Kejia Li
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China
| | - Yau-Tsz Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China
| | - Hon-Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Yi-Wah Mak
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Hao Yao
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopaedic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, China
| | - Jing Sun
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China
| | - Michael Tim-Yun Ong
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China
| | - Kevin Ki-Wai Ho
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
| | - Chien-Wei Lee
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Oscar Kuang-Sheng Lee
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
| | - Patrick Shu-Hang Yung
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China
| | - Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| |
Collapse
|
33
|
Xu K, Zhang J, Ma W, Wang Y, Chen B, Gao N, Pang J, Zhan H. Home-Based Shi's Knee Daoyin Exercise for Knee Osteoarthritis: A Randomized Controlled Pilot Trial. J Pain Res 2024; 17:2811-2822. [PMID: 39224147 PMCID: PMC11368107 DOI: 10.2147/jpr.s469176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Objective Shi's Knee Daoyin (SKD) exercise is a treatment derived from Traditional Chinese exercise (TCE) specifically designed for lower limb health care. This study aimed to assess the feasibility of conducting a randomized controlled trial to explore the effectiveness of SKD exercise in treating knee osteoarthritis (KOA). Methods Participants were randomized to receive Health Education (HE) or SKD exercise. The primary outcomes were feasibility and safety outcomes, including participant recruitment rate, retention rate, as well as adherence to intervention. The secondary outcomes included Visual Analogue Scale (VAS) scores for pain, the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score, the 20-Meter Walk Test (20-MWT) and the 5-times Chair-Stand Test (5-CST). Results The results indicate that out of 89 individuals invited to participate in the study, 72 were eligible and agreed to participate, resulting in a recruitment rate of 80.9%. All participating patients completed the follow-up and were included in the analysis; no patients dropped out of the study due to adverse events. The secondary outcome measures showed that after twelve weeks of treatment, the VAS score, WOMAC total score, WOMAC pain score, WOMAC stiffness score, and WOMAC function score of patients in the HE group and SKD group all improved, but the improvement was more significant in the SKD group. The 20-MWT of SKD group after treatment was significantly shorter than before treatment (P<0.001); There was no significant difference in 20-MWT between the HE group and baseline after treatment. The performance of the two groups of patients improved in 5-CST, but there was no statistical difference between the two groups after treatment (P=2.439). Conclusion This study evaluated the feasibility and effectiveness of home-based SKD exercise intervention in alleviating symptoms in patients with symptomatic KOA, providing valuable information for designing an appropriate randomized controlled study.
Collapse
Affiliation(s)
- Kun Xu
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jiefan Zhang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Wei Ma
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yongyu Wang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Bo Chen
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Ningyang Gao
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jian Pang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Hongsheng Zhan
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
34
|
Zhang Y, Huang W, Xiao H, Ruan S, Deng J. NGF-BMSC-SF/CS composites for repairing knee joint osteochondral defects in rabbits: evaluation of the repair effect and potential underlying mechanisms. J Orthop Surg Res 2024; 19:443. [PMID: 39075502 PMCID: PMC11285204 DOI: 10.1186/s13018-024-04801-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/20/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND With the rapid growth of the ageing population, chronic diseases such as osteoarthritis have become one of the major diseases affecting the quality of life of elderly people. The main pathological manifestation of osteoarthritis is articular cartilage damage. Alleviating and repairing damaged cartilage has always been a challenge. The application of cartilage tissue engineering methods has shown promise for articular cartilage repair. Many studies have used cartilage tissue engineering methods to repair damaged cartilage and obtained good results, but these methods still cannot be used clinically. Therefore, this study aimed to investigate the effect of incorporating nerve growth factor (NGF) into a silk fibroin (SF)/chitosan (CS) scaffold containing bone marrow-derived mesenchymal stem cells (BMSCs) on the repair of articular cartilage defects in the knees of rabbits and to explore the possible underlying mechanism involved. MATERIALS AND METHODS Nerve growth factor-loaded sustained-release microspheres were prepared by a double emulsion solvent evaporation method. SF/CS scaffolds were prepared by vacuum drying and chemical crosslinking. BMSCs were isolated and cultured by density gradient centrifugation and adherent culture. NGF-SF/CS-BMSC composites were prepared and implanted into articular cartilage defects in the knees of rabbits. The repair of articular cartilage was assessed by gross observation, imaging and histological staining at different time points after surgery. The repair effect was evaluated by the International Cartilage Repair Society (ICRS) score and a modified Wakitani score. In vitro experiments were also performed to observe the effect of different concentrations of NGF on the proliferation and directional differentiation of BMSCs on the SF/CS scaffold. RESULTS In the repair of cartilage defects in rabbit knees, NGF-SF/CS-BMSCs resulted in higher ICRS scores and lower modified Wakitani scores. The in vitro results showed that there was no significant correlation between the proliferation of BMSCs and the addition of different concentrations of NGF. Additionally, there was no significant difference in the protein and mRNA expression of COL2a1 and ACAN between the groups after the addition of different concentrations of NGF. CONCLUSION NGF-SF/CS-BMSCs improved the repair of articular cartilage defects in the knees of rabbits. This repair effect may be related to the early promotion of subchondral bone repair.
Collapse
Affiliation(s)
- Yong Zhang
- The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi City, Guizhou Province, 563000, China
- The People's Hospital of Bozhou District, Zunyi City, Guizhou Province, 563000, China
| | - Wenliang Huang
- The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi City, Guizhou Province, 563000, China
| | - Hongli Xiao
- The First People's Hospital of Guiyang City, Guiyang, Guizhou Province, 550002, China
| | - Shiqiang Ruan
- The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi City, Guizhou Province, 563000, China
| | - Jiang Deng
- The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi City, Guizhou Province, 563000, China.
| |
Collapse
|
35
|
Kou H, Ma J. Network pharmacology prediction and molecular docking-based strategy to explore the potential mechanism of Duhuo-Jisheng pair against osteoarthritis. J Pharm Pharmacol 2024; 76:743-752. [PMID: 38183673 DOI: 10.1093/jpp/rgad117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/12/2023] [Indexed: 01/08/2024]
Abstract
OBJECTIVE The Duhuo-Jisheng pair is the ruling herb in Duhuo Jisheng decoction, which is a classic formula first recorded in the preparedness and urgency of the thousand jewels. METHODS We obtained the primary constituents of Duhuo-Jisheng and their associated protein targets from the TCMSP database. We constructed a composite target network using Cytoscape 3.9.1. To identify potential targets for the treatment of osteoarthritis (OA), we retrieved disease targets from OMIM and GeneCards databases and compared them with the composite targets. We imported the overlapping targets into the STRING database to construct a protein-protein interaction (PPI) network. We also conducted Gene ontology (GO) and KEGG enrichment analyses on the targets. RESULTS The component target network consisted of numerous nodes and edges. Notably, quercetin, ammidin, and β-sitosterol were identified as the compounds with high degrees. The PPI network identified tumour necrosis factor (TNF), TP53, and NOS2 as proteins with high degrees. The results of GO and KEGG analyses revealed that the signalling pathways used by DHQJD to treat OA included the NF-κB, PI3K-AKT, and TNF pathways. CONCLUSION Our study provides insights into the effective components and potential molecular mechanisms of Duhuo-Jisheng in treating OA, thus serving as a reference for further basic research in this field.
Collapse
Affiliation(s)
- Haiyang Kou
- Department of Joint Surgery, Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi, China
| | - Jianbing Ma
- Department of Joint Surgery, Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi, China
| |
Collapse
|
36
|
Lee M, Lee H, Chung H, Lee JH, Kim D, Cho S, Kim TJ, Kim HS. Micro-current stimulation could inhibit IL-1β-induced inflammatory responses in chondrocytes and protect knee bone cartilage from osteoarthritis. Biomed Eng Lett 2024; 14:801-812. [PMID: 38946809 PMCID: PMC11208348 DOI: 10.1007/s13534-024-00376-1] [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: 12/18/2023] [Revised: 02/24/2024] [Accepted: 03/29/2024] [Indexed: 07/02/2024] Open
Abstract
This study aimed to evaluate the inhibitory effects of micro-current stimulation (MCS) on inflammatory responses in chondrocytes and degradation of extracellular matrix (ECM) in osteoarthritis (OA). To determine the efficacy of MCS, IL-1β-treated chondrocytes and monosodium iodoacetate (MIA)-induced OA rat model were used. To evaluate the cytotoxicity and nitric oxide (NO) production in SW1353 cells, the presence or absence of IL-1β treatment or various levels of MCS were applied. Immunoblot analysis was conducted to evaluate whether MCS can modulate IL-1R1/MyD88/NF-κB signaling pathway and various indicators involved in ECM degradation. Additionally, to determine whether MCS alleviates subchondral bone structure destruction caused by OA, micro-CT analysis, immunoblot analysis, and ELISA were conducted using OA rat model. 25 and 50 µA levels of MCS showed effects in cell proliferation and NO production. The MCS group with IL-1β treatment lead to significant inhibition of protein expression levels regarding IL-1R1/MyD88/NF-κB signaling and reduction of the nucleus translocation of NF-κB. In addition, the protein expression levels of MMP-1, MMP-3, MMP-13, and IL-1β decreased, whereas collagen II and aggrecan increased. In animal results, morphological analysis of subchondral bone using micro-CT showed that MCS induced subchondral bone regeneration and improvement, as evidenced by increased thickness and bone mineral density of the subchondral bone. Furthermore, MCS-applied groups showed decreases in the protein expression of MMP-1 and MMP-3, while increases in collagen-II and aggrecan expressions. These findings suggest that MCS has the potential to be used as a non-pharmaceutical method to alleviate OA.
Collapse
Affiliation(s)
- Minjoo Lee
- Department of Biomedical Engineering, Yonsei University, Gangwon, 26493 South Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Gangwon, 26493 South Korea
| | - Halim Chung
- Department of Biomedical Engineering, Yonsei University, Gangwon, 26493 South Korea
| | - Jin-Ho Lee
- Division of Biological Science and Technology, Yonsei University, Gangwon, 26493 South Korea
| | - Doyong Kim
- Department of Biomedical Engineering, Yonsei University, Gangwon, 26493 South Korea
| | | | - Tack-Joong Kim
- Division of Biological Science and Technology, Yonsei University, Gangwon, 26493 South Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Gangwon, 26493 South Korea
| |
Collapse
|
37
|
Liang Q, Cheng Z, Qin L. Advanced nanoparticles in osteoarthritis treatment. BIOMATERIALS TRANSLATIONAL 2024; 5:95-113. [PMID: 39351157 PMCID: PMC11438607 DOI: 10.12336/biomatertransl.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/21/2024] [Accepted: 04/11/2024] [Indexed: 10/04/2024]
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disorder, affecting hundreds of millions of people globally. Current clinical approaches are confined to providing only symptomatic relief. Research over the past two decades has established that OA is not merely a process of wear and tear of the articular cartilage but involves abnormal remodelling of all joint tissues. Although many new mechanisms of disease have been identified in the past several decades, the efficient and sustainable delivery of drugs targeting these mechanisms in joint tissues remains a major challenge. Nanoparticles recently emerged as favoured delivery vehicles in OA treatment, offering extended drug retention, enhanced drug targeting, and improved drug stability and solubility. In this review, we consider OA as a disease affecting the entire joint and initially explore the pathophysiology of OA across multiple joint tissues, including the articular cartilage, synovium, fat pad, bone, and meniscus. We then classify nanoparticles based on their composition and structure, such as lipids, polymers, inorganic materials, peptides/proteins, and extracellular vesicles. We summarise the recent advances in their use for treatment and diagnosis of OA. Finally, we discuss the current challenges and future directions in this field. In conclusion, nanoparticle-based nanosystems are promising carriers that advance OA treatment and diagnosis.
Collapse
Affiliation(s)
- Qiushi Liang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
38
|
Sarig-Rapaport H, Krupnik S, Rowan TG. Amorphous calcium carbonate as a novel potential treatment for osteoarthritis in dogs: a pilot clinical study. Front Vet Sci 2024; 11:1381941. [PMID: 38983767 PMCID: PMC11231089 DOI: 10.3389/fvets.2024.1381941] [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: 02/04/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024] Open
Abstract
Background Amorphous calcium carbonate (ACC) is a potential new treatment for canine osteoarthritis (OA) with novel mechanisms based on local pH modulation and targeting bone remodeling, inflammation, and pain. The aim of this pilot exploratory clinical study was to obtain initial data on the potential efficacy and safety of ACC in OA dogs and to determine if further investigation was appropriate using similar assessment methods. Materials and methods In this prospective, randomized, double-blind, controlled pilot study, 41 client-owned dogs were allocated in a 2:1 ratio to ACC: placebo given orally for 56 days. Efficacy assessments included improvements in pain and mobility using owner questionnaires [Canine Brief Pain Inventory (CBPI), Client Specific Outcome Measure (CSOM), and Veterinary Orthopedic Scores (VOS)]. Safety in the study population was monitored by veterinary examinations, clinical pathology, and adverse events. Results Fifty-three dogs were screened, of which 41 enrolled and served for the safety assessment. Thirty-six dogs were found evaluable for initial efficacy assessment. Three dogs given placebo (21.4%) and one given ACC (4.5%) were removed before day 56 due to owner-perceived pain and were considered treatment failures. There were no serious adverse events or clinically significant treatment-related effects in the study. Overall, ACC was found safe in the small study population. On day 56, proportionally more ACC than placebo dogs were treatment successes based on CBPI (45.5% vs. 21.4%) and CSOM (63.6% vs. 30.8%, respectively); however, these differences were not statistically significant (p = 0.15 and 0.06, respectively). On day 56, within the ACC group but not the placebo group, the CBPI, CSOM, and VOS assessments were lower compared to day 0 and day 14 (p < 0.05). Limitations The relatively small number of dogs limited the statistical power of the pilot study in evaluating the efficacy and safety of ACC. Conclusion Study results support the conduct of larger, appropriately powered studies using similar assessments to confirm whether ACC may be a safe and effective treatment for OA in dogs.
Collapse
|
39
|
Faeed M, Ghiasvand M, Fareghzadeh B, Taghiyar L. Osteochondral organoids: current advances, applications, and upcoming challenges. Stem Cell Res Ther 2024; 15:183. [PMID: 38902814 PMCID: PMC11191177 DOI: 10.1186/s13287-024-03790-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024] Open
Abstract
In the realm of studying joint-related diseases, there is a continuous quest for more accurate and representative models. Recently, regenerative medicine and tissue engineering have seen a growing interest in utilizing organoids as powerful tools for studying complex biological systems in vitro. Organoids, three-dimensional structures replicating the architecture and function of organs, provide a unique platform for investigating disease mechanisms, drug responses, and tissue regeneration. The surge in organoid research is fueled by the need for physiologically relevant models to bridge the gap between traditional cell cultures and in vivo studies. Osteochondral organoids have emerged as a promising avenue in this pursuit, offering a better platform to mimic the intricate biological interactions within bone and cartilage. This review explores the significance of osteochondral organoids and the need for their development in advancing our understanding and treatment of bone and cartilage-related diseases. It summarizes osteochondral organoids' insights and research progress, focusing on their composition, materials, cell sources, and cultivation methods, as well as the concept of organoids on chips and application scenarios. Additionally, we address the limitations and challenges these organoids face, emphasizing the necessity for further research to overcome these obstacles and facilitate orthopedic regeneration.
Collapse
Affiliation(s)
- Maryam Faeed
- Cell and Molecular School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mahsa Ghiasvand
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahar Fareghzadeh
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Leila Taghiyar
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
40
|
Feng K, Wang F, Chen H, Zhang R, Liu J, Li X, Xie X, Kang Q. Cartilage progenitor cells derived extracellular vesicles-based cell-free strategy for osteoarthritis treatment by efficient inflammation inhibition and extracellular matrix homeostasis restoration. J Nanobiotechnology 2024; 22:345. [PMID: 38890638 PMCID: PMC11186174 DOI: 10.1186/s12951-024-02632-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease which currently lacks of effective agents. It is therefore urgent and necessary to seek an effective approach that can inhibit inflammation and promote cartilage matrix homeostasis. Cartilage progenitor cells (CPCs) are identified as a cell population of superficial zone in articular cartilage which possess strong migration ability, proliferative capacity, and chondrogenic potential. Recently, the application of CPCs may represent a novel cell therapy strategy for OA treatment. There is growing evidence that extracellular vesicles (EVs) are primary mediators of the benefits of stem cell-based therapy. In this study, we explored the protective effects of CPCs-derived EVs (CPCs-EVs) on IL-1β-induced chondrocytes. We found CPCs-EVs exhibited chondro-protective effects in vitro. Furthermore, our study demonstrated that CPCs-EVs promoted matrix anabolism and inhibited inflammatory response at least partially via blocking STAT3 activation. In addition, liquid chromatography-tandem mass spectrometry analysis identified 991 proteins encapsulated in CPCs-EVs. By bioinformatics analysis, we showed that STAT3 regulatory proteins were enriched in CPCs-EVs and could be transported to chondrocytes. To promoting the protective function of CPCs-EVs in vivo, CPCs-EVs were modified with cationic peptide ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD) for surface charge reverse. In posttraumatic OA mice, our results showed PPD modified CPCs-EVs (PPD-EVs) effectively inhibited extracellular matrix catabolism and attenuated cartilage degeneration. Moreover, PPD-EVs down-regulated inflammatory factors expressions and reduced OA-related pain in OA mice. In ex-vivo cultured OA cartilage explants, PPD-EVs successfully promoted matrix anabolism and inhibited inflammation. Collectively, CPCs-EVs-based cell-free therapy is a promising strategy for OA treatment.
Collapse
Affiliation(s)
- Kai Feng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Feng Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Hongfang Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Rui Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jiashuo Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaodong Li
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xuetao Xie
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Qinglin Kang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
41
|
Chu M, Chen G, Chen K, Zhu P, Wang Z, Qian Z, Tao H, Xu Y, Geng D. Heme oxygenase 1 linked to inactivation of subchondral osteoclasts in osteoarthritis. J Zhejiang Univ Sci B 2024; 25:513-528. [PMID: 38910496 PMCID: PMC11199094 DOI: 10.1631/jzus.b2300303] [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: 05/08/2023] [Accepted: 08/09/2023] [Indexed: 06/25/2024]
Abstract
Osteoarthritis (OA) is a chronic progressive osteoarthropathy in the elderly. Osteoclast activation plays a crucial role in the occurrence of subchondral bone loss in early OA. However, the specific mechanism of osteoclast differentiation in OA remains unclear. In our study, gene expression profiles related to OA disease progression and osteoclast activation were screened from the Gene Expression Omnibus (GEO) repository. GEO2R and Funrich analysis tools were employed to find differentially expressed genes (DEGs). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses demonstrated that chemical carcinogenesis, reactive oxygen species (ROS), and response to oxidative stress were mainly involved in osteoclast differentiation in OA subchondral bone. Furthermore, fourteen DEGs that are associated with oxidative stress were identified. The first ranked differential gene, heme oxygenase 1 (HMOX1), was selected for further validation. Related results showed that osteoclast activation in the pathogenesis of OA subchondral bone is accompanied by the downregulation of HMOX1. Carnosol was revealed to inhibit osteoclastogenesis by targeting HMOX1 and upregulating the expression of antioxidant protein in vitro. Meanwhile, carnosol was found to alleviate the severity of OA by inhibiting the activation of subchondral osteoclasts in vivo. Our research indicated that the activation of osteoclasts due to subchondral bone redox dysplasia may serve as a significant pathway for the advancement of OA. Targeting HMOX1 in subchondral osteoclasts may offer novel insights for the treatment of early OA.
Collapse
Affiliation(s)
- Miao Chu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, Yixing People's Hospital, Yixing 214200, China
| | - Guangdong Chen
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Kai Chen
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, Hai'an People's Hospital, Hai'an 226600, China
| | - Pengfei Zhu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zhen Wang
- Department of Orthopedics, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou 215028, China
| | - Zhonglai Qian
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Huaqiang Tao
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China. ,
| | - Yaozeng Xu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Dechun Geng
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
42
|
Feng J, Zhang Q, Pu F, Zhu Z, Lu K, Lu WW, Tong L, Yu H, Chen D. Signalling interaction between β-catenin and other signalling molecules during osteoarthritis development. Cell Prolif 2024; 57:e13600. [PMID: 38199244 PMCID: PMC11150147 DOI: 10.1111/cpr.13600] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/29/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent disorder of synovial joint affecting multiple joints. In the past decade, we have witnessed conceptual switch of OA pathogenesis from a 'wear and tear' disease to a disease affecting entire joint. Extensive studies have been conducted to understand the underlying mechanisms of OA using genetic mouse models and ex vivo joint tissues derived from individuals with OA. These studies revealed that multiple signalling pathways are involved in OA development, including the canonical Wnt/β-catenin signalling and its interaction with other signalling pathways, such as transforming growth factor β (TGF-β), bone morphogenic protein (BMP), Indian Hedgehog (Ihh), nuclear factor κB (NF-κB), fibroblast growth factor (FGF), and Notch. The identification of signalling interaction and underlying mechanisms are currently underway and the specific molecule(s) and key signalling pathway(s) playing a decisive role in OA development need to be evaluated. This review will focus on recent progresses in understanding of the critical role of Wnt/β-catenin signalling in OA pathogenesis and interaction of β-catenin with other pathways, such as TGF-β, BMP, Notch, Ihh, NF-κB, and FGF. Understanding of these novel insights into the interaction of β-catenin with other pathways and its integration into a complex gene regulatory network during OA development will help us identify the key signalling pathway of OA pathogenesis leading to the discovery of novel therapeutic strategies for OA intervention.
Collapse
Affiliation(s)
- Jing Feng
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of WuhanTongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Department of OrthopedicsWuhan No. 1 HospitalWuhanHubeiChina
| | - Qing Zhang
- Department of EmergencyRenmin Hospital, Wuhan UniversityWuhanHubeiChina
| | - Feifei Pu
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of WuhanTongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Department of OrthopedicsWuhan No. 1 HospitalWuhanHubeiChina
| | - Zhenglin Zhu
- Department of Orthopedic Surgerythe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Ke Lu
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
- Research Center for Computer‐aided Drug DiscoveryShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - William W. Lu
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
| | - Liping Tong
- Research Center for Computer‐aided Drug DiscoveryShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Huan Yu
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of WuhanTongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Department of OrthopedicsWuhan No. 1 HospitalWuhanHubeiChina
| | - Di Chen
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
- Research Center for Computer‐aided Drug DiscoveryShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| |
Collapse
|
43
|
Xu L, Ma J, Yu Q, Zhu K, Wu X, Zhou C, Lin X. Evidence supported by Mendelian randomization: impact on inflammatory factors in knee osteoarthritis. Front Med (Lausanne) 2024; 11:1382836. [PMID: 38863887 PMCID: PMC11165061 DOI: 10.3389/fmed.2024.1382836] [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: 02/11/2024] [Accepted: 05/01/2024] [Indexed: 06/13/2024] Open
Abstract
Background Prior investigations have indicated associations between Knee Osteoarthritis (KOA) and certain inflammatory cytokines, such as the interleukin series and tumor necrosis factor-alpha (TNFα). To further elaborate on these findings, our investigation utilizes Mendelian randomization to explore the causal relationships between KOA and 91 inflammatory cytokines. Methods This two-sample Mendelian randomization utilized genetic variations associated with KOA from a large, publicly accessible Genome-Wide Association Study (GWAS), comprising 2,227 cases and 454,121 controls of European descent. The genetic data for inflammatory cytokines were obtained from a GWAS summary involving 14,824 individuals of European ancestry. Causal relationships between exposures and outcomes were primarily investigated using the inverse variance weighted method. To enhance the robustness of the research results, other methods were combined to assist, such as weighted median, weighted model and so on. Multiple sensitivity analysis, including MR-Egger, MR-PRESSO and leave one out, was also carried out. These different analytical methods are used to enhance the validity and reliability of the final results. Results The results of Mendelian randomization indicated that Adenosine Deaminase (ADA), Fibroblast Growth Factor 5(FGF5), and Hepatocyte growth factor (HFG) proteins are protective factors for KOA (IVWADA: OR = 0.862, 95% CI: 0.771-0.963, p = 0.008; IVWFGF5: OR = 0.850, 95% CI: 0.764-0.946, p = 0.003; IVWHFG: OR = 0.798, 95% CI: 0.642-0.991, p = 0.042), while Tumor necrosis factor (TNFα), Colony-stimulating factor 1(CSF1), and Tumor necrosis factor ligand superfamily member 12(TWEAK) proteins are risk factors for KOA. (IVWTNFα: OR = 1.319, 95% CI: 1.067-1.631, p = 0.011; IVWCSF1: OR = 1.389, 95% CI: 1.125-1.714, p = 0.002; IVWTWEAK: OR = 1.206, 95% CI: 1.016-1.431, p = 0.032). Conclusion The six proteins identified in this study demonstrate a close association with the onset of KOA, offering valuable insights for future therapeutic interventions. These findings contribute to the growing understanding of KOA at the microscopic protein level, paving the way for potential targeted therapeutic approaches.
Collapse
Affiliation(s)
- Lilei Xu
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaqi Ma
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Yu
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kean Zhu
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuewen Wu
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuanlong Zhou
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Acupuncture, Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianming Lin
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Acupuncture, Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
44
|
Zhang G, Qin J, Xu W, Liu M, Wu R, Qin Y. Gene expression and immune infiltration analysis comparing lesioned and preserved subchondral bone in osteoarthritis. PeerJ 2024; 12:e17417. [PMID: 38827307 PMCID: PMC11141552 DOI: 10.7717/peerj.17417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/28/2024] [Indexed: 06/04/2024] Open
Abstract
Background Osteoarthritis (OA) is a degenerative disease requiring additional research. This study compared gene expression and immune infiltration between lesioned and preserved subchondral bone. The results were validated using multiple tissue datasets and experiments. Methods Differentially expressed genes (DEGs) between the lesioned and preserved tibial plateaus of OA patients were identified in the GSE51588 dataset. Moreover, functional annotation and protein-protein interaction (PPI) network analyses were performed on the lesioned and preserved sides to explore potential therapeutic targets in OA subchondral bones. In addition, multiple tissues were used to screen coexpressed genes, and the expression levels of identified candidate DEGs in OA were measured by quantitative real-time polymerase chain reaction. Finally, an immune infiltration analysis was conducted. Results A total of 1,010 DEGs were identified, 423 upregulated and 587 downregulated. The biological process (BP) terms enriched in the upregulated genes included "skeletal system development", "sister chromatid cohesion", and "ossification". Pathways were enriched in "Wnt signaling pathway" and "proteoglycans in cancer". The BP terms enriched in the downregulated genes included "inflammatory response", "xenobiotic metabolic process", and "positive regulation of inflammatory response". The enriched pathways included "neuroactive ligand-receptor interaction" and "AMP-activated protein kinase signaling". JUN, tumor necrosis factor α, and interleukin-1β were the hub genes in the PPI network. Collagen XI A1 and leucine-rich repeat-containing 15 were screened from multiple datasets and experimentally validated. Immune infiltration analyses showed fewer infiltrating adipocytes and endothelial cells in the lesioned versus preserved samples. Conclusion Our findings provide valuable information for future studies on the pathogenic mechanism of OA and potential therapeutic and diagnostic targets.
Collapse
Affiliation(s)
- Gang Zhang
- The Second Affiliated Hospital of Harbin Medical University, Department of Orthopedics Surgery, Harbin Medical University, Harbin, China
- Department of Orthopedics, Harbin First Hospital, Harbin, China
- Future Medicine Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinwei Qin
- Department of Emergency, Harbin First Hospital, Harbin, China
| | - Wenbo Xu
- The Second Affiliated Hospital of Harbin Medical University, Department of Orthopedics Surgery, Harbin Medical University, Harbin, China
| | - Meina Liu
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Rilige Wu
- Medical Big Data Research Center, Medical Innovation Research Division of PLA General Hospital, Beijing, China
| | - Yong Qin
- The Second Affiliated Hospital of Harbin Medical University, Department of Orthopedics Surgery, Harbin Medical University, Harbin, China
| |
Collapse
|
45
|
Xie Y, Shao F, Ji Y, Feng D, Wang L, Huang Z, Wu S, Sun F, Jiang H, Miyamoto A, Wang H, Zhang C. Network Analysis of Osteoarthritis Progression Using a Steiner Minimal Tree Algorithm. J Inflamm Res 2024; 17:3201-3209. [PMID: 38779430 PMCID: PMC11110812 DOI: 10.2147/jir.s438407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 12/09/2023] [Indexed: 05/25/2024] Open
Abstract
Purpose To provide a comprehensive analysis of associated genes with osteoarthritis (OA). Here, we reported a network analysis of OA progression by using a Steiner minimal tree algorithm. Methods We collected the OA-related genes through screening the publications in MEDLINE. We performed functional analysis to analyze the associated biochemical pathways of the OA-related genes. Pathway crosstalk analysis was constructed to explore interactions of the enriched pathways. Steiner minimal tree algorithm was used to analyze molecular pathway networks. The average clustering coefficient was compared with the corresponding values of the Osteoarthritis-specific network. The new finding RNA was compared with former single-cell RNA-seq analysis results. Results A gene set with 177 members reported to be significantly associated with Osteoarthritis was collected from 187 studies. Functional enrichment analysis revealed a specific related-OA gene including skeletal system development, cytokine-mediated signaling pathway, inflammatory response, cartilage development, and extracellular matrix organization. We performed a pathway crosstalk analysis among the 72 significantly enriched pathways. A total of 151 of the 177 genes in the Osteoarthritis gene set were included in the human interactome network. There were 31 genes in the former single-cell RNA-seq analysis results. The CLU, ENO1, SRRM1, UBC, HMGB1, NR3C1, NOTCH2NL, and CBX5 have significantly increased expression in seven molecularly defined populations of OA cartilage. Conclusion The Steiner tree-based approach finds new biological molecules associated with OA genes.
Collapse
Affiliation(s)
- Yujie Xie
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Fanglin Shao
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yuxiu Ji
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Dechao Feng
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Ling Wang
- School of Information and Communication Engineering, University of Electronic Science and Technology, Chengdu, Sichuan, People’s Republic of China
| | - Zonghai Huang
- School of Information and Communication Engineering, University of Electronic Science and Technology, Chengdu, Sichuan, People’s Republic of China
| | - Shengjian Wu
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Fuhua Sun
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Hong Jiang
- Rehabilitation Medicine Department, Xichong County People’s hospital, Nanchong, Sichuan, People’s Republic of China
| | - Akira Miyamoto
- Faculty of Rehabilitation, Nishikyushu University, Kansaitama, Japan
| | - Haiming Wang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Chi Zhang
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
46
|
Yokota S, Ishizu H, Miyazaki T, Takahashi D, Iwasaki N, Shimizu T. Osteoporosis, Osteoarthritis, and Subchondral Insufficiency Fracture: Recent Insights. Biomedicines 2024; 12:843. [PMID: 38672197 PMCID: PMC11048726 DOI: 10.3390/biomedicines12040843] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The increased incidence of osteoarthritis (OA), particularly knee and hip OA, and osteoporosis (OP), owing to population aging, have escalated the medical expense burden. Osteoarthritis is more prevalent in older women, and the involvement of subchondral bone fragility spotlights its association with OP. Notably, subchondral insufficiency fracture (SIF) may represent a more pronounced condition of OA pathophysiology. This review summarizes the relationship between OA and OP, incorporating recent insights into SIF. Progressive SIF leads to joint collapse and secondary OA and is associated with OP. Furthermore, the thinning and fragility of subchondral bone in early-stage OA suggest that SIF may be a subtype of OA (osteoporosis-related OA, OPOA) characterized by significant subchondral bone damage. The high bone mineral density observed in OA may be overestimated due to osteophytes and sclerosis and can potentially contribute to OPOA. The incidence of OPOA is expected to increase along with population aging. Therefore, prioritizing OP screening, early interventions for patients with early-stage OA, and fracture prevention measures such as rehabilitation, fracture liaison services, nutritional management, and medication guidance are essential.
Collapse
Affiliation(s)
| | | | | | | | | | - Tomohiro Shimizu
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (S.Y.); (H.I.); (T.M.); (D.T.); (N.I.)
| |
Collapse
|
47
|
Farinelli L, Riccio M, Gigante A, De Francesco F. Pain Management Strategies in Osteoarthritis. Biomedicines 2024; 12:805. [PMID: 38672160 PMCID: PMC11048725 DOI: 10.3390/biomedicines12040805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Pain is the major symptom of osteoarthritis (OA) and is an important factor in strategies to manage this disease. However, the current standard of care does not provide satisfactory pain relief for many patients. The pathophysiology of OA is complex, and its presentation as a clinical syndrome is associated with the pathologies of multiple joint tissues. Treatment options are generally classified as pharmacologic, nonpharmacologic, surgical, and complementary and/or alternative, typically used in combination to achieve optimal results. The goals of treatment are the alleviation of symptoms and improvement in functional status. Several studies are exploring various directions for OA pain management, including tissue regeneration techniques, personalized medicine, and targeted drug therapies. The aim of the present narrative review is to extensively describe all the treatments available in the current practice, further describing the most important innovative therapies. Advancements in understanding the molecular and genetic aspects of osteoarthritis may lead to more effective and tailored treatment approaches in the future.
Collapse
Affiliation(s)
- Luca Farinelli
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60121 Ancona, Italy; (L.F.); (A.G.)
| | - Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, Azienda Ospedaliera Universitaria delle Marche, 60126 Ancona, Italy;
| | - Antonio Gigante
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60121 Ancona, Italy; (L.F.); (A.G.)
| | - Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, Azienda Ospedaliera Universitaria delle Marche, 60126 Ancona, Italy;
| |
Collapse
|
48
|
Li X, Chen W, Liu D, Chen P, Wang S, Li F, Chen Q, Lv S, Li F, Chen C, Guo S, Yuan W, Li P, Hu Z. Pathological progression of osteoarthritis: a perspective on subchondral bone. Front Med 2024; 18:237-257. [PMID: 38619691 DOI: 10.1007/s11684-024-1061-y] [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: 11/21/2023] [Accepted: 01/17/2024] [Indexed: 04/16/2024]
Abstract
Osteoarthritis (OA) is a degenerative bone disease associated with aging. The rising global aging population has led to a surge in OA cases, thereby imposing a significant socioeconomic burden. Researchers have been keenly investigating the mechanisms underlying OA. Previous studies have suggested that the disease starts with synovial inflammation and hyperplasia, advancing toward cartilage degradation. Ultimately, subchondral-bone collapse, sclerosis, and osteophyte formation occur. This progression is deemed as "top to bottom." However, recent research is challenging this perspective by indicating that initial changes occur in subchondral bone, precipitating cartilage breakdown. In this review, we elucidate the epidemiology of OA and present an in-depth overview of the subchondral bone's physiological state, functions, and the varied pathological shifts during OA progression. We also introduce the role of multifunctional signal pathways (including osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL)/receptor activator of nuclear factor-kappa B (RANK), and chemokine (CXC motif) ligand 12 (CXCL12)/CXC motif chemokine receptor 4 (CXCR4)) in the pathology of subchondral bone and their role in the "bottom-up" progression of OA. Using vivid pattern maps and clinical images, this review highlights the crucial role of subchondral bone in driving OA progression, illuminating its interplay with the condition.
Collapse
Affiliation(s)
- Xuefei Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Wenhua Chen
- Research and Development Center of Chinese Medicine Resources and Biotechnology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan Liu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pinghua Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shiyun Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangfang Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qian Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shunyi Lv
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangyu Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Chen Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Suxia Guo
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Weina Yuan
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pan Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhijun Hu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
49
|
Vlashi R, Zhang X, Li H, Chen G. Potential therapeutic strategies for osteoarthritis via CRISPR/Cas9 mediated gene editing. Rev Endocr Metab Disord 2024; 25:339-367. [PMID: 38055160 DOI: 10.1007/s11154-023-09860-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
Osteoarthritis (OA) is an incapacitating and one of the most common physically degenerative conditions with an assorted etiology and a highly complicated molecular mechanism that to date lacks an efficient treatment. The capacity to design biological networks and accurately modify existing genomic sites holds an apt potential for applications across medical and biotechnological sciences. One of these highly specific genomes editing technologies is the CRISPR/Cas9 mechanism, referred to as the clustered regularly interspaced short palindromic repeats, which is a defense mechanism constituted by CRISPR associated protein 9 (Cas9) directed by small non-coding RNAs (sncRNA) that bind to target DNA through Watson-Crick base pairing rules where subsequent repair of the target DNA is initiated. Up-to-date research has established the effectiveness of the CRISPR/Cas9 mechanism in targeting the genetic and epigenetic alterations in OA by suppressing or deleting gene expressions and eventually distributing distinctive anti-arthritic properties in both in vitro and in vivo osteoarthritic models. This review aims to epitomize the role of this high-throughput and multiplexed gene editing method as an analogous therapeutic strategy that could greatly facilitate the clinical development of OA-related treatments since it's reportedly an easy, minimally invasive technique, and a comparatively less painful method for osteoarthritic patients.
Collapse
Affiliation(s)
- Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, China.
- Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
50
|
Li M, Teng M, Mao G, Sun Y, Liu S, Li R, Wang X, Qiu Y. PUD@HA/PEEK Scaffold Induces Subchondral Bone Regeneration to Repair Osteochondral Defect in Rabbits. ACS Biomater Sci Eng 2024; 10:1006-1017. [PMID: 38252806 DOI: 10.1021/acsbiomaterials.3c01352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Osteochondral defects (OCDs) pose a significant challenge in clinical practice, and recent advancements in their repair indicate that satisfying subchondral bone repair may be critical for this. Herein, a series of hydroxyapatite/poly(ether ether ketone) (HA/PEEK) scaffolds were fabricated with varying mass percentages (0, 20, 30, and 40%) to induce subchondral bone regeneration. Subsequently, an optimal scaffold with 40% HA/PEEK was selected to establish osteochondral scaffolds with poly(ether urethane) urea-Danshensu (PUD) for repairing the OCD. The material characteristics of HA/PEEK and PUD were investigated using scanning electron microscopy, tensile, swelling, and fatigue tests, and cytological experiments. The effects of serial HA/PEEK scaffolds on subchondral bone repair were then assessed by using microcomputed tomography, hard tissue slicing, and histological staining. Furthermore, the optimal 40% HA/PEEK scaffold was used to develop osteochondral scaffolds with PUD to observe the effect on the OCD repair. HA/PEEK materials exhibited an even HA distribution in PEEK. However, when composited with HA, PEEK exhibited inferior mechanical strength. 40%HA/PEEK scaffolds showed an optimum effect on in vivo subchondral bone repair. Cartilage regeneration on 40%HA/PEEK scaffolds was pronounced. After PUD was introduced onto the HA/PEEK, the PUD@40%HA/PEEK scaffold produced the expected effect on the repair of the OCD in rabbits. Therefore, achieving satisfactory subchondral bone repair can benefit surficial cartilage repair. The PUD@40%HA/PEEK scaffold could induce subchondral bone regeneration to repair the OCD in rabbits and could provide a novel approach for the repair of the OCD in clinical practice.
Collapse
Affiliation(s)
- Meng Li
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061 Shaanxi, China
| | - Menghao Teng
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061 Shaanxi, China
| | - Genwen Mao
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yiqing Sun
- The Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shenghang Liu
- The Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Run Li
- The Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiangyu Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yusheng Qiu
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061 Shaanxi, China
| |
Collapse
|