1
|
Zheng G, Lu M, Ouyang Y, Sun G. RNA methylation: A new perspective in osteoarthritis research. Gene 2025; 959:149518. [PMID: 40254081 DOI: 10.1016/j.gene.2025.149518] [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: 02/04/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by cartilage degradation, osteophyte formation, and joint dysfunction, significantly impairing the quality of life in the elderly population. Recently, RNA modifications, as a dynamic and reversible epigenetic modification, have emerged as critical players in the onset and progression of OA. This review systematically summarizes the major types of RNA modifications involved in OA, including N6-methyladenosine (m6A), 5-methylcytosine (m5C), and 7-methylguanosine (m7G), and explores their roles in regulating chondrocyte autophagy, inflammatory responses, and key signaling pathways. with a primary focus on RNA methylation. Special emphasis is placed on the dynamic regulatory functions of key methyltransferases (e.g., METTL3, FTO, WTAP) and their potential contributions to OA pathogenesis. Furthermore, we address current research hotspots and controversies in the field, proposing future research directions, such as leveraging single-cell sequencing to decipher dynamic RNA modification changes during OA progression and uncovering the cooperative networks among various RNA modifications. Advancing our understanding of the biological roles and mechanisms of RNA modifications holds promise for innovative strategies in the early diagnosis, disease stratification, and targeted therapy of OA.
Collapse
Affiliation(s)
- Guihao Zheng
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China; Graduate School of Jiangxi Medical College, Nanchang University, China.
| | - Meifeng Lu
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China; Graduate School of Jiangxi Medical College, Nanchang University, China.
| | - Yulong Ouyang
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China.
| | - Guicai Sun
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China.
| |
Collapse
|
2
|
Dong L, Yang Z, Liu J, Wu R, Liao Y, Kuang L. SERPINF1 knockdown attenuates chondrocyte senescence, hypertrophy, and inflammation in osteoarthritis to offer a potential therapeutic strategy. Cell Signal 2025; 132:111840. [PMID: 40306348 DOI: 10.1016/j.cellsig.2025.111840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/15/2025] [Accepted: 04/26/2025] [Indexed: 05/02/2025]
Abstract
Osteoarthritis (OA) is characterized by cartilage degradation, synovial inflammation, subchondral bone remodeling, and osteophyte formation, leading to chronic pain and impaired mobility. Chondrocyte senescence, inflammation, and hypertrophic differentiation critically contribute to OA progression. Integrated analysis of four GEO datasets identified SERPINF1 as a consistently upregulated gene in both human and animal OA samples. Histopathological and immunohistochemical analyses confirmed increased SERPINF1 in OA cartilage, where chondrocytes showed elevated SERPINF1 protein alongside reduced aggrecan expression. Functional studies revealed that SERPINF1 knockdown in OA chondrocytes diminished senescence markers (p21, p16, p53) while increasing Lamin B1, and reduced levels of pro-inflammatory cytokines (IL-1β, TNF-α, and IL-6). Conversely, overexpression of SERPINF1 in normal chondrocytes induced senescence and increased inflammatory mediator expression, accompanied by altered extracellular matrix metabolism and hypertrophy marker expression. Mechanistic analysis further implicated the TNF-α/NF-κB signaling pathway in mediating these effects. In a destabilization of the medial meniscus (DMM) mouse model, intra-articular SERPINF1 knockdown attenuated cartilage destruction, reduced senescence and inflammatory markers, and restored ECM integrity. Collectively, these findings demonstrate that SERPINF1 promotes OA progression by exacerbating chondrocyte senescence, inflammation, and hypertrophy, suggesting that targeting SERPINF1 may offer a novel therapeutic strategy for OA.
Collapse
Affiliation(s)
- Lini Dong
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Zhiwei Yang
- Department of Orthopaedics, Changde Hospital of Xiangya School of Medicine, Central South University (The First People's Hospital of Changde), Changde 415000, Hunan, China
| | - Jie Liu
- Department of Spinal Surgery, The Fourth People's Hospital of Guiyang, Guiyang 550002, Guizhou, China
| | - Ren Wu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yunlong Liao
- Department of Spinal Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Lei Kuang
- Department of Spinal Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
3
|
Jacob J, Aggarwal A, Bhattacharyya S, Sahni D, Sharma V, Aggarwal A. Fisetin and resveratrol exhibit senotherapeutic effects and suppress cellular senescence in osteoarthritic cartilage-derived chondrogenic progenitor cells. Eur J Pharmacol 2025; 997:177573. [PMID: 40189080 DOI: 10.1016/j.ejphar.2025.177573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
Chondrogenic progenitor cells (CPCs) in the articular cartilage of knee osteoarthritis (OA) patients exhibit cellular senescence and its associated secretory phenotype (SASP). We hypothesized that the senescence of CPCs can be suppressed using natural compounds. This study aimed to evaluate the senotherapeutic effects of fisetin and resveratrol to suppress the cellular senescence in CPCs. In vitro, pre-treatment of CPCs with increasing doses of fisetin and resveratrol (5μM-100μM) were non-cytotoxic, decreased the senescence index and dampened the expression of cellular senescence markers, p53 and p38MAPK. Additionally, SASP-related genes and proteins (MMP-9, MMP13) and inflammatory mediators (IL-1β, TGF-β, and IL-6) were downregulated. Further, in silico analysis confirmed the high binding affinity of these natural drugs to OA-related proteins. Overall, fisetin and resveratrol dampened the senescence of CPCs by downregulating the p53 effector protein and effectively reducing the SASP. From this study, natural compound candidates proved to be potential drug candidates that suppress senescence via p53.
Collapse
Affiliation(s)
- Justin Jacob
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Aditya Aggarwal
- Department of Orthopedics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Daisy Sahni
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Vinit Sharma
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Anjali Aggarwal
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India.
| |
Collapse
|
4
|
Mahrous MN, Attia EZ, Abdelkader MSA, Elsayed Abouzed DE, Gebril SM, Alnajjar R, Al-Karmalawy AA, Hamed ANE. Chemical composition and anti-osteoarthritis potential of Lolium perenne L. assisted with computational studies. Fitoterapia 2025; 183:106551. [PMID: 40252739 DOI: 10.1016/j.fitote.2025.106551] [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: 02/03/2025] [Revised: 04/05/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Osteoarthritis (OA) is a serious health issue that lacks a totally effective treatment. In light of the critical need for alternative therapies to halt osteoarthritis and its progress, This study explores the potential of Lolium perenne as a treatment for osteoarthritis for the first time. The research evaluates the plant's protective effects against monosodium iodoacetate (MIA)-induced OA in rats. 24 rats were assigned into 4 groups, group I (sham control), group II (OA), group III (dichloromethane fraction (FrII) + OA), and group IV (ethyl acetate fraction (FrIII) + OA). For 21 days, the samples were given orally at a dose of 40 mg/kg. The obtained results indicated all treatment groups showed a significant decrease in the severity of articular cartilage degradation and in the concentration of Interleukin-1 beta (IL-1β) and Tumor necrosis factor alpha (TNF-α) cytokines compared to the OA-induced group Moreover, eight structurally varied metabolites were isolated and identified from the FrII and FrIII fractions derived from the methanol extract of the aerial part of L. perenne, which were linked with pro-inflammatory cytokines in the computational docking investigation. Interestingly, salcolin B (2b) and calquiquelignan E (4) isolated from FrII fraction, showed prevalent activity for the two tested receptors [salcolin B, IL-1β (-6.7354 kcal/mol) and calquiquelignan E, TNF-α (-6.2038 kcal/mol)] compared to the reference flurbiprofen drug. Finally, the frontier candidates against IL-1β and TNF-α receptors simulated at 100 ns and the MM-GBSA calculations confirming the docking results. These findings highlight L. perenne's potential as a natural therapeutic for OA.
Collapse
Affiliation(s)
| | - Eman Zekry Attia
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, Minia National University, New Minia, Egypt
| | | | - Deiaa E Elsayed Abouzed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| | - Sahar M Gebril
- Department of Histology and Cell biology, Faculty of Medicine, Sohag University, Sohag 82524, Egypt
| | - Radwan Alnajjar
- Department of Chemistry, Faculty of Science, University of Benghazi, Benghazi, Libya
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad 10023, Iraq; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Ashraf Nageeb Elsayed Hamed
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, Minia National University, New Minia, Egypt.
| |
Collapse
|
5
|
Ooi K, Yamamoto K, Kobayashi Y, Javaheri B, Jensen A, Kanakis I, Sakai T, Jarad F, Nakamura H, Pitsillides AA, Kawashiri S, Bou-Gharios G. Temporomandibular joint degeneration arises spontaneously in STR/ort mice and is prevented by targeted aggrecanase inhibition. OSTEOARTHRITIS AND CARTILAGE OPEN 2025; 7:100599. [PMID: 40207030 PMCID: PMC11981737 DOI: 10.1016/j.ocarto.2025.100599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
OBJECTIVE Temporomandibular joint osteoarthritis (TMJ-OA) is painful and causes masticatory dysfunction, but current treatment is limited to symptom relief due to an incomplete appreciation of aetiology. Herein, we develop morphological and histological methods for quantitative evaluation of TMJ-OA severity and examine whether STR/Ort mice, which are genetically predisposed to spontaneous knee OA, exhibit protection against TMJ-OA upon genetic gain-of-function modification of an aggrecanase-selective mutant of tissue inhibitor of metalloproteinase (TIMP)-3. DESIGN We established morphological changes in mandibular condylar head adapted from human TMJ-OA criteria, and developed and verified the utility of TMJ-OA histological damage scoring adapted from the OARSI system. Mutant TIMP3 containing an extra alanine at the N-Terminus ([-1A] TIMP-3 was overexpressed in STR/Ort and CBA mice. Morphological changes in mandibular condyle and TMJ cartilage degradation were evaluated and quantified using micro-CT and histology in mice aged 10, 20 and 40 weeks. RESULTS Whilst no evidence of TMJ-OA was observed in STR/Ort mice aged 10 weeks, bone erosion and osteophyte formation appeared in the mandibular condyle by 20 weeks, with remarkable deformity and bone resorption at 40 weeks in STR/Ort, but not the parental CBA strain. TMJ-OA was less severe in 40 week-old [-1A]TIMP-3 overexpressing STR/Ort and CBA compared to wild-type mice. CONCLUSIONS Using our new mouse TMJ-OA scoring system we have found that OA affects joints other than the knee in the STR/Ort strain. Genetic gain-of-function modification of STR/Ort mice with an aggrecanase-selective mutant of tissue inhibitor of metalloproteinase (TIMP)-3 also affords in vivo chondroprotection against this TMJ-OA.
Collapse
Affiliation(s)
- Kazuhiro Ooi
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Kazuhiro Yamamoto
- Department of Musculoskeletal and Aging Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Yutaka Kobayashi
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Behzad Javaheri
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Anders Jensen
- Department of Musculoskeletal and Aging Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Ioannis Kanakis
- Chester Medical School, Faculty of Medicine and Life Sciences, University of Chester, Chester, United Kingdom
| | - Takao Sakai
- Department of Diagnostic Pathology, Faculty of Medicine, Fujita Health University, Aichi, Japan
| | - Fadi Jarad
- Department of School of Dentistry, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Hiroyuki Nakamura
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Science, Ryukyu University, Okinawa, Japan
| | - Andrew A. Pitsillides
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Shuichi Kawashiri
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - George Bou-Gharios
- Department of Musculoskeletal and Aging Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
6
|
Conceição F, Meneses J, Lebre F, Becker M, Araújo-Gomes N, Vos R, Ribeiro AR, Alfaro-Moreno E, Leijten J, Moreira Teixeira L. Sex-stratified osteochondral organ-on-chip model reveals sex-specific responses to inflammatory stimulation. Mater Today Bio 2025; 32:101728. [PMID: 40242482 PMCID: PMC12000750 DOI: 10.1016/j.mtbio.2025.101728] [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: 02/03/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Osteoarthritis (OA) is a musculoskeletal degenerative disease characterized by alterations in cartilage and subchondral bone leading to impaired joint function. OA disproportionally affects females more than males, yet the molecular mechanisms underlying these biological sex differences remain elusive. Current therapeutic strategies to halt the progression of OA are still lacking, in part due to the limited predictive potential of standard models which often do not account for sex disparities. Herein, an organ-on-chip microfluidic platform was developed to model the osteochondral unit, composed of adjacent bone and cartilage culture chambers, and capture sex-specific hallmarks of OA. Sex-stratified human primary chondrocytes and osteoblasts were compartmentalized within biomimetic hydrogels emulating the bone-cartilage interface, which were subjected to inflammatory triggers to mimic the onset of OA. We confirmed that interleukin-1β and Tumor Necrosis Factor-α stimulation triggered upregulation of pro-inflammatory cytokines and matrix metalloproteinases related genes in all donors, with marginal trends for increased expression in female cells. In addition, metabolic labeling coupled with confocal imaging revealed that inflammatory stimulation modulated extracellular matrix deposition by human chondrocytes in a sex-specific fashion. Not only matrix deposition but also matrix remodeling was altered upon inflammation, leading to a significant reduction in matrix stiffness in both cartilage and bone compartments. Overall, sex-stratified osteochondral unit on-chips offer novel insights into sex-specific cellular responses to inflammatory insults, demonstrating the importance of incorporating sex stratification in emergent organ-on-chip models. Thus, this platform provides a physiologically relevant 3D microenvironment to further investigate sex-specific drivers of OA, paving the way for targeted therapies.
Collapse
Affiliation(s)
- Francisco Conceição
- Department of Bioengineering Technologies, Faculty of Science and Technology, TechMedCentre, University of Twente, 7522 NB, Enschede, the Netherlands
| | - João Meneses
- Department of Bioengineering Technologies, Faculty of Science and Technology, TechMedCentre, University of Twente, 7522 NB, Enschede, the Netherlands
| | - Filipa Lebre
- Nanosafety Group, International Iberian Nanotechnology Laboratory, 4715-330, Braga, Portugal
| | - Malin Becker
- Optics11 Life, 1101 BM, Amsterdam, the Netherlands
| | - Nuno Araújo-Gomes
- Department of Bioengineering Technologies, Faculty of Science and Technology, TechMedCentre, University of Twente, 7522 NB, Enschede, the Netherlands
| | - Rianne Vos
- Department of Bioengineering Technologies, Faculty of Science and Technology, TechMedCentre, University of Twente, 7522 NB, Enschede, the Netherlands
| | - Ana R. Ribeiro
- Nanosafety Group, International Iberian Nanotechnology Laboratory, 4715-330, Braga, Portugal
| | - Ernesto Alfaro-Moreno
- Nanosafety Group, International Iberian Nanotechnology Laboratory, 4715-330, Braga, Portugal
| | - Jeroen Leijten
- Department of Bioengineering Technologies, Faculty of Science and Technology, TechMedCentre, University of Twente, 7522 NB, Enschede, the Netherlands
| | - Liliana Moreira Teixeira
- Department of Bioengineering Technologies, Faculty of Science and Technology, TechMedCentre, University of Twente, 7522 NB, Enschede, the Netherlands
- Organ-on-Chip Centre Twente, TechMed Centre, MESA+, University of Twente, 7522 NB, Enschede, the Netherlands
| |
Collapse
|
7
|
Guo H, Wang S, Zhang Y, Sun J, Guo L, Pang J, Zhan H. α2-Macroglobulin Promotes Chondrocyte Proliferation and Cartilage Matrix Synthesis via Inducing PCNA. Cartilage 2025; 16:202-211. [PMID: 37872706 PMCID: PMC12066844 DOI: 10.1177/19476035231207776] [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: 06/04/2023] [Revised: 09/17/2023] [Accepted: 09/29/2023] [Indexed: 10/25/2023] Open
Abstract
Objectivesα2-Macroglobulin (A2M) can prevent cartilage degeneration by blocking many types of cartilage-degrading enzymes, but the mechanism remains to be clarified. This study aimed to test that A2M protects against cartilage degeneration by promoting chondrocyte proliferation and cartilage matrix synthesis via inducing proliferating cell nuclear antigen (PCNA).DesignThe cartilage degeneration of the anterior cruciate ligament transection (ACLT) model was evaluated by Safranin O-fast green staining, and articular cartilage degeneration was graded using the Osteoarthritis Research Society International (OARSI)-modified Mankin criteria. The chondrocyte proliferation was detected by 5-Bromodeoxyuridinc (BrdU), MTT, and Cell Counting Kit-8 (CCK8) methods. The chondrocyte apoptosis was detected by lactate dehydrogenase (LDH) assay and Annexin PI staining with the flow cytometer. The glycosaminoglycan (sGAG) and aggrecan in culture supernatant were measured by enzyme-linked immunosorbent assay (ELISA). Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to analyze the type II collagen and aggrecan mRNA expression. The PCNA protein expression was analyzed by western blot and immunofluorescent staining.ResultsA2M can attenuate cartilage degeneration in ACLT rats. The OARSI scores for cartilage degeneration in the A2M group were lower than those in the phosphate-buffered saline (PBS) group. A2M can promote chondrocyte proliferation and inhibit chondrocyte apoptosis, promote the cartilage matrix synthesis in chondrocytes (type II collagen and aggrecan), and culture supernatant (sGAG and aggrecan). At the same time, it also up-regulated the PCNA protein expression in chondrocytes.ConclusionsA2M can promote chondrocyte proliferation and cartilage matrix synthesis via inducing PCNA expression.
Collapse
Affiliation(s)
- Hailing Guo
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shaowei Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University; Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Yang Zhang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University; Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Jian Sun
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University; Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Li Guo
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University; Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Jian Pang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Hongsheng Zhan
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
8
|
Konola VM, Multanen J, Ihalainen JK, Hintikka JE, Jämsä T, Kautiainen H, Nieminen MT, Pekkala S, Valtonen M, Heinonen A. Effects of high impact exercise on systemic cytokines in women with mild knee osteoarthritis: A 12-month RCT. OSTEOARTHRITIS AND CARTILAGE OPEN 2025; 7:100609. [PMID: 40290652 PMCID: PMC12033985 DOI: 10.1016/j.ocarto.2025.100609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/30/2025] [Indexed: 04/30/2025] Open
Abstract
Objective This study investigated the effects of a high-impact exercise regimen compared with a reference group on systemic cytokine levels in patients with mild knee osteoarthritis (OA). Furthermore, associations between cytokines and magnetic resonance imaging (MRI) transverse relaxation time (T2) mapping and metabolic equivalent task hours (MET-hours) during leisure-time physical activity (LTPA) were assessed. Method In this secondary analysis, 73 postmenopausal women aged 50-65 years with mild knee OA were randomized to a 12-month high-impact aerobic/step aerobics training group (n = 35) or a non-training reference group (n = 38). The serum cytokine levels, including interleukin-1 alpha (IL-1α), IL-2, IL-4, IL-5, IL-6, IL-10, IL-13, IL-17, interferon-gamma (IFN-γ), and tumor necrosis factor alpha (TNF-α), were determined via multiplex cytokine assays. The cartilage structure of the medial tibial condyle was assessed by MRI T2 mapping. The primary outcome was between-group differences in cytokine level changes. Results After a 12-month follow-up, no significant differences in cytokine level changes were found between the groups. In the intervention group, 12-month changes in TNF-α levels were associated with changes in medial tibial condyle T2. In the reference group, 12-month changes in IL-10 levels were associated with changes in medial tibial condyle T2 and the number of weekly LTPA MET-hours. Conclusion A progressive high-impact exercise regimen did not affect systemic cytokine levels compared to the reference group and could therefore offer a possible mode of exercise for postmenopausal women with mild knee OA. Trial registration number ISRCTN58314639.
Collapse
Affiliation(s)
- Ville-Markus Konola
- Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35, Jyväskylä, FI, 40014, Finland
| | - Juhani Multanen
- South-Eastern Finland University of Applied Sciences, Savonlinna, Finland
| | - Johanna K. Ihalainen
- Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35, Jyväskylä, FI, 40014, Finland
| | - Jukka E. Hintikka
- Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35, Jyväskylä, FI, 40014, Finland
| | - Timo Jämsä
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Hannu Kautiainen
- Primary Health Care Unit, Kuopio University Hospital, Kuopio, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Miika T. Nieminen
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Diagnostics, Oulu University Hospital, Oulu, Finland
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35, Jyväskylä, FI, 40014, Finland
| | - Maarit Valtonen
- Finnish Institute of High Performance Sport KIHU, Jyväskylä, Finland
| | - Ari Heinonen
- Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35, Jyväskylä, FI, 40014, Finland
| |
Collapse
|
9
|
Du D, Zhong H, Huang Z, Gao M, Su R, Xu M, Shi L, Hu J, Cao H. Polysaccharides isolated from shufeng jiedu capsules by cross-flow ultrafiltration show anti-inflammatory effects on LPS-stimulated RAW264.7 cells and zebrafish inflammatory models. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119817. [PMID: 40250639 DOI: 10.1016/j.jep.2025.119817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shufeng Jiedu Capsules (SFJDC) is a traditional Chinese patent medicine comprising eight traditional Chinese medicines (TCM). SFJDC is known for its anti-inflammatory and antipyretic effects and is mainly used in clinics to treat upper respiratory tract infections. Currently, studies on the active ingredients of the SFJDC all focus on small-molecule compounds. In contrast, bio-macromolecules, such as the anti-inflammatory activities of polysaccharides in SFJDC, have not been studied, and the composition of the polysaccharides in SFJDC is also unclear. AIM OF THE STUDY This study aimed to isolate active polysaccharides from Shufeng Jiedu capsules and determine their structural properties and anti-inflammatory activities. MATERIALS AND METHODS The polysaccharides with different molecular weights were prepared by organic solvent extraction, alcohol precipitation, dialysis, and cross-flow ultrafiltration. The structural characterization of polysaccharides was clarified by high-performance size exclusion chromatography (HPGPC), ion chromatography (IC), and Fourier transform infrared spectroscopy (FT-IR). Enzyme-linked immunosorbent assay (ELISA) and quantitative real-time PCR (qRT-PCR) assay were used to investigate the anti-inflammatory effects of polysaccharides on Lipopolysaccharides (LPS)-stimulated RAW264.7 cells. The in vivo study was employed on the CuSO4-induced and LPS-stimulated zebrafish inflammatory models, and the survival analysis, observation of neutrophil migration, hematoxylin-eosin (H&E) staining, and qRT-PCR assays were used to investigate the in vivo anti-inflammatory effect of polysaccharides. RESULTS The crude polysaccharides SFJDC-CP were obtained from the mixed aqueous extract of SFJDC with a yield of 38.72 %. Both SFJDC and SFJDC-CP dose-dependently inhibited the secretion of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-1β in LPS-stimulated RAW264.7 cells, and SFJDC-CP was more effective at a lower dosage. SFJDC-CP was further separated into three fractions, SFJDC-CP1-CP3, by cross-flow ultrafiltration apparatus with nominal molecular weight cut-offs of 100 kDa, 50 kDa, and 10 kDa membrane cassettes, and the yields were approximately 58.19 %, 10.88 %, and 30.94 %, respectively. The MWs of the SFJDC-CP and its SFJDC-CP1-CP3 were 35.7 kDa, 149.1 kDa, 34.5 kDa, and 15.1 kDa, respectively. The four polysaccharides were composed of rhamnose, arabinose, galactose, glucose, and galacturonic acid in different molar ratios. Non-toxic concentrations of the four polysaccharides ranged from 12.5 to 200 μg/mL. The four polysaccharides significantly reduced the mRNA expression levels and release of IL-1β, IL-6, and TNF-α (P < 0.0001) in LPS-stimulated RAW264.7 cells. Polysaccharides also decreased inflammatory cell infiltration and neutrophil migration (P < 0.05 or P < 0.001) in both CuSO4-induced and LPS-microinjected zebrafish inflammatory models. Additionally, they effectively inhibited the mRNA levels of IL-6 and TNF-α in LPS-infected zebrafish (P < 0.01 or P < 0.001). CONCLUSIONS Polysaccharides isolated from Shufeng Jiedu capsules have demonstrated anti-inflammatory effects on LPS-stimulated RAW264.7 cells and zebrafish inflammatory models. This study provided preliminary evidence that polysaccharides are one of the main anti-inflammatory ingredients of Shufeng Jiedu capsules. Additionally, it may provide valuable perspectives for investigating polysaccharides in other TCM formulations, particularly those obtained through aqueous extraction methods.
Collapse
Affiliation(s)
- Dongsheng Du
- College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, PR China.
| | - Hongjiao Zhong
- College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, PR China
| | - Ziyi Huang
- Traditional Chinese Pharmacological Laboratory, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China
| | - Mingzhu Gao
- College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, PR China
| | - Ruirui Su
- College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, PR China
| | - Mengqiu Xu
- College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, PR China
| | - Lei Shi
- Wannan Medical College, Wuhu, 241002, Anhui, PR China
| | - Jie Hu
- Wuhu Food and Drug Inspection Center, Wuhu 241008, Anhui, PR China
| | - Huihui Cao
- Traditional Chinese Pharmacological Laboratory, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China.
| |
Collapse
|
10
|
Mei Z, Yilamu K, Ni W, Shen P, Pan N, Chen H, Su Y, Guo L, Sun Q, Li Z, Huang D, Fang X, Fan S, Zhang H, Shen S. Chondrocyte fatty acid oxidation drives osteoarthritis via SOX9 degradation and epigenetic regulation. Nat Commun 2025; 16:4892. [PMID: 40425566 DOI: 10.1038/s41467-025-60037-4] [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: 07/22/2024] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Osteoarthritis is the most prevalent age-related degenerative joint disease and is closely linked to obesity. However, the underlying mechanisms remain unclear. Here we show that altered lipid metabolism in chondrocytes, particularly enhanced fatty acid oxidation (FAO), contributes to osteoarthritis progression. Excessive FAO causes acetyl-CoA accumulation, thereby altering protein-acetylation profiles, where the core FAO enzyme HADHA is hyperacetylated and activated, reciprocally boosting FAO activity and exacerbating OA progression. Mechanistically, elevated FAO reduces AMPK activity, impairs SOX9 phosphorylation, and ultimately promotes its ubiquitination-mediated degradation. Additionally, acetyl-CoA orchestrates epigenetic modulation, affecting multiple cellular processes critical for osteoarthritis pathogenesis, including the transcriptional activation of MMP13 and ADAMTS7. Cartilage-targeted delivery of trimetazidine, an FAO inhibitor and AMPK activator, demonstrates superior efficacy in a mouse model of metabolism-associated post-traumatic osteoarthritis. These findings suggest that targeting chondrocyte-lipid metabolism may offer new therapeutic strategies for osteoarthritis.
Collapse
Affiliation(s)
- Zixuan Mei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Kamuran Yilamu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Weiyu Ni
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Panyang Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Nan Pan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Huasen Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Yingfeng Su
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Lei Guo
- Pooling Institute of Translational Medicine, Hangzhou, China
| | - Qunan Sun
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhaomei Li
- Department of Geriatrics, Xiaoshan Geriatric Hospital, Hangzhou, China
| | - Dongdong Huang
- Pooling Institute of Translational Medicine, Hangzhou, China
| | - Xiangqian Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Haitao Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
11
|
ZhaYang XZ, Chen YX, Hua WD, Bai ZL, Jin YP, Zhao XW, Liu QF, Meng ZD. Integrating bioinformatics and machine learning to identify biomarkers of branched chain amino acid related genes in osteoarthritis. BMC Musculoskelet Disord 2025; 26:517. [PMID: 40420260 DOI: 10.1186/s12891-025-08779-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/20/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Branched-chain amino acids (BCAA) metabolism is significantly associated with osteoarthritis (OA), but the specific mechanism of BCAA related genes (BCAA-RGs) in OA is still unclear. Therefore, this research intended to identify potential biomarkers and mechanisms of action of BCAA-RGs in OA tissues. METHODS Differential genes were obtained from the Gene Expression Omnibus (GEO) database and intersections were taken with BCAA-RGs to identify candidate genes. The underlying mechanisms were revealed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Subsequently, by combining three machine learning algorithms to identify genes with highly correlated OA features. In addition, created diagnostic maps and subject Receiver operating characteristic curves (ROCs) to assess the ability of the signature genes to diagnose OA and to predict their possible roles in molecular regulatory network axes and molecular signaling pathways. RESULTS Eight candidate genes were acquired by intersecting 4,178 DEGs and 14 BCAA-RGs. Subsequently, five candidate biomarkers were obtained, namely SLC3A2, SLC7A5, SLC43A2, SLC43A1, and SLC7A7. Importantly, SLC3A2 and SLC7A5 were validated by validation set and qRT-PCR. Furthermore, the nomogram constructed by SLC3A2 and SLC7A5 exhibited excellent accuracy in predicting the incidence of OA. The enrichment results demonstrated that SLC3A2 and SLC7A5 were significantly enriched in ribosome, insulin signaling pathway, olfactory transduction, etc. Meanwhile, we also found XIST regulated SLC7A5 through hsa-miR-30e-5p, and regulated SLC3A2 through hsa-miR-7-5p.OIP5-AS1 regulated SLC7A5 and SLC3A2 through hsa-miR-7-5p. By the way, 150 drugs were identified, including Acetaminophen and Acrylamide, which exhibited simultaneous targeting of these two biomarkers. CONCLUSION Based on bioinformatics, SLC3A2 and SLC7A5 were identified as biomarkers related to BCAA in OA, which may provide a new reference for the treatment and diagnosis of OA patients.
Collapse
Affiliation(s)
- Xiao-Zhi ZhaYang
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yan-Xiong Chen
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Wen-Da Hua
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zheng-Lin Bai
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yun-Peng Jin
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xing-Wen Zhao
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Quan-Fu Liu
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zeng-Dong Meng
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Digital Orthopedics, Kunming, Yunnan, China.
- Department of Orthopedic Surgery in The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, No.157, Jinbi Road, Xishan District, Kunming, Yunnan, 650032, China.
| |
Collapse
|
12
|
Shao Z, Wang T, Yan X, Ning R, Xu X, He Q, Zhang X, Jiang M, Yang C. Identification of a RANKL/TNF-α Dual-Inhibitor as a Potential Disease-Modifying Agent for the Treatment of Knee Osteoarthritis. J Med Chem 2025; 68:10216-10237. [PMID: 40358029 DOI: 10.1021/acs.jmedchem.5c00394] [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: 05/15/2025]
Abstract
Osteoarthritis (OA) is a multifactorial degenerative disease involved subchondral bone remodeling, cartilage destruction and synovium inflammation. While receptor activator of nuclear factor-κB ligand (RANKL), a tumor necrosis factor (TNF) superfamily protein, is the critical regulator in bone metabolism associated with subchondral bone resorption, TNF-α is also an important inflammatory factor involved in the OA inflammation and cartilage destruction. Based on previous compound Y1599, we identified a novel tetrahydro-β-carboline derivative Y2641 with both RANKL and TNF-α inhibition in this study. Y2641 exhibited potent RANKL-induced osteoclastogenic inhibition (IC50 = 109.1 nM), and had anti-inflammatory and cartilage destruction inhibiting effects at 10 μM with low cytotoxicity. SPR assays demonstrated the binding affinity of Y2641 to RANKL (Kd = 3.984 μM) and TNF-α (Kd = 18.59 μM). In vivo assay further revealed the disease-modifying effects of Y2641 in OA rats, establishing Y2641 as a promising lead compound for the development of disease-modifying osteoarthritis drugs.
Collapse
Affiliation(s)
- Zhengguang Shao
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 200021, China
| | - Tianqi Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xueming Yan
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruonan Ning
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xing Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian He
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaofei Zhang
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Min Jiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chunhao Yang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 200021, China
| |
Collapse
|
13
|
Gao C, Pu H, Zeng Y, Xiao J. The causal relationship between immune cells and knee osteoarthritis: Mendelian randomization study. BMC Musculoskelet Disord 2025; 26:504. [PMID: 40405089 PMCID: PMC12096777 DOI: 10.1186/s12891-025-08735-4] [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: 12/17/2023] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
Knee osteoarthritis (OA) is a common degenerative joint disease that affects millions of people worldwide. Inflammation is one of the key pathogenic factors of knee OA. However, the causal relationship between immune cells and knee OA development remains unclear. Herein, we used Mendelian randomization (MR) analysis to evaluate causal relationship between 731 immune cells and knee OA. Several methods were applied to ensure the robustness of our results, including inverse-variance weighted (IVW), simple mode, weighted median, weighted mode, and MR-Egger. We found that 23 immune cell phenotypes were causally associated with knee OA (P < 0.05), including various subpopulations of B cells, T cells, TBNK (T cells, B cells, Natural Killer cells) and monocytes, which was confirmed by heterogeneity, sensitivity, and pleiotropy tests. B cells had dominant effects on OA development, and specifically, our findings suggest that BAFF-R in IgD + CD38- unswitched memory B cells may have a protective role, whereas CD25 in IgD + CD24 + B cells appears to be associated with increased risk, pending further validation. Moreover, a higher population of regulatory T (Treg) cells indicated a higher risk of OA and reversely, OA could induce Treg differentiation. Collectively, our study identified several immune cells that were closely related to OA development, which provided novel insights into the pathogenesis of OA and therapeutic targets for OA treatment.
Collapse
Affiliation(s)
- Chenghao Gao
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, People's Republic of China
| | - Hongxu Pu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, People's Republic of China
| | - Yifan Zeng
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, People's Republic of China
| | - Jun Xiao
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, People's Republic of China.
| |
Collapse
|
14
|
Pawlikowski M, Sikora S, Ostrowski G. The role of mathematical models in prediction of osteoarthritis development. Comput Biol Med 2025; 193:110407. [PMID: 40403629 DOI: 10.1016/j.compbiomed.2025.110407] [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: 08/01/2024] [Revised: 04/17/2025] [Accepted: 05/17/2025] [Indexed: 05/24/2025]
Abstract
In the paper we presented the review of mathematical and numerical models of osteoarthritis (OA). As angiogenesis seems to be the most principal factor in OA mathematical and numerical modelling, we focused on the models that consider the process. The spectrum of the presented models is wide. They were divided in the scale of the simulated phenomena, i.e., micro- or macro-scale. A part of them considers only damage of tissue without paying attention to its remodeling. Others consider loss of tissue, new tissue formulation and remodeling of bone, both in micro- and macro-scale. What is worth mentioning is that most of the models were confirmed by comparing results to data available in literature. Only a few of them were experimentally validated. As the conclusion, we stated that the most accurate models are those that take into consideration mechanical stimulation, biological signaling and their nonlocal effects. Also, an important feature of an OA model is the ability to adapt it to various cases to be able to simulate OA in any joint.
Collapse
Affiliation(s)
- Marek Pawlikowski
- Institute of Mechanics and Printing, Warsaw University of Technology, Ul. Narbutta 85, 02-524, Warszawa, Poland.
| | - Szymon Sikora
- Institute of Mechanics and Printing, Warsaw University of Technology, Ul. Narbutta 85, 02-524, Warszawa, Poland
| | - Gustaw Ostrowski
- Institute of Mechanics and Printing, Warsaw University of Technology, Ul. Narbutta 85, 02-524, Warszawa, Poland
| |
Collapse
|
15
|
Lv Q, Zhao X, Teng S, Jin X, Zhou Y, Sun Y, Pei H, Yan Z, Ma C. DNA Origami-Based CD44-Targeted Therapy Silences Stat3 Enhances Cartilage Regeneration and Alleviates Osteoarthritis Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e03939. [PMID: 40396977 DOI: 10.1002/advs.202503939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/10/2025] [Indexed: 05/22/2025]
Abstract
Osteoarthritis (OA) is a widespread musculoskeletal disorder affecting ≈600 million people globally, and small interfering RNA (siRNA) therapy shows potential in targeting OA progression. However, the efficient and targeted delivery of siRNA remains a major challenge due to issues with tissue specificity and degradation in vivo. In this study, A DNA origami-based chondrocyte-targeted delivery system (OCS) is designed for siRNA delivery to OA-affected cartilage. The DNA origami is engineered to load with siRNA targeting signal transducer and activator of transcription 3 (Stat3), a key regulator of inflammation and cartilage degradation, and is functionalized with anti-CD44 aptamers for selective targeting of OA chondrocytes. In vitro, the DNA origami system effectively delivers siRNA to diseased chondrocytes, silencing matrix metalloproteinases expression and reducing inflammation. In OA rat models, it preserves cartilage integrity, promotes regeneration, and mitigates ECM degradation without evident side effects. These findings highlight DNA origami as a promising platform for siRNA-based OA therapy, offering a promising solution to the challenges of targeted and efficient siRNA delivery.
Collapse
Affiliation(s)
- Qi Lv
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xiang Zhao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Songsong Teng
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, 215009, China
- Joint Laboratory of Biomaterials and Translational Medicine, Puheng Technology Co., Ltd, Suzhou, 215000, China
| | - Xinmeng Jin
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Ying Zhou
- Shanghai Key laboratory of green chemistry and chemical Processes, School of chemistry and Molecular engineering, Shanghai center of Brain inspired intelligent Materials and devices, East China normal University, Shanghai, 200241, China
| | - Yueyang Sun
- Shanghai Key laboratory of green chemistry and chemical Processes, School of chemistry and Molecular engineering, Shanghai center of Brain inspired intelligent Materials and devices, East China normal University, Shanghai, 200241, China
| | - Hao Pei
- Shanghai Key laboratory of green chemistry and chemical Processes, School of chemistry and Molecular engineering, Shanghai center of Brain inspired intelligent Materials and devices, East China normal University, Shanghai, 200241, China
| | - Zuoqin Yan
- Institute of Bone and Joint Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
- Department of Orthopedic Surgery, Shanghai Geriatric Medical Center, Shanghai, 201104, China
| | - Chunhui Ma
- Institute of Bone and Joint Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
- Department of Orthopedic Surgery, Shanghai Geriatric Medical Center, Shanghai, 201104, China
| |
Collapse
|
16
|
Yang C, Liao X, Wang Y, Zhong W, Xian F, Gao M, Zeng M, Chen J, Chen X, Li M, Zhou K. Biomimetic Nanoplatform Integrating CeO₂ Nanozymes and Anti-Inflammatory Peptides for Osteoarthritis Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2502677. [PMID: 40391633 DOI: 10.1002/smll.202502677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/13/2025] [Indexed: 05/22/2025]
Abstract
Osteoarthritis (OA) affects more than 250 million people worldwide, with current therapies focused primarily on symptom management rather than addressing underlying disease mechanisms. Here, MHTCK, a novel biomimetic nanoplatform is presented that uniquely integrates CeO₂ nanozymes with the anti-inflammatory peptide KAFAK through the fusion of a macrophage-synoviocyte membrane coating. Physicochemical characterization reveals that MHTCK maintains nanostability with a size of ≈ 200 nm and a surface charge of -30 mV. Compared with conventional antioxidant nanoparticles, the platform demonstrated superior cellular uptake in synoviocytes (2.8-fold), chondrocytes (3.2-fold), and macrophages (4.1-fold). In vitro studies revealed that MHTCK effectively scavenged multiple ROS species by targeting mitochondria while preserving mitochondrial function as evidenced by the maintenance of ATP production, and promotion of M2 macrophage polarization. The biomimetic membrane coating enabled prolonged joint retention of up to 10 days postinjection through specific tissue-targeting mechanisms, significantly improving pain thresholds and cartilage preservation in an OA rat model. This work demonstrates how rational integration of nanozyme technology with peptide therapeutics in a biomimetic delivery system can effectively modulate both oxidative stress and inflammation in OA, while maintaining cellular bioenergetics, providing new insights for developing targeted nanotherapeutics for inflammatory joint diseases.
Collapse
Affiliation(s)
- Chengli Yang
- Department of Pharmacy, Clinical Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Hospital of Guizhou Medical University, Guizhou, 550004, P. R. China
| | - Xukun Liao
- Department of Pharmacy, Clinical Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Hospital of Guizhou Medical University, Guizhou, 550004, P. R. China
| | - Yilin Wang
- Department of Pharmacy, Clinical Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Hospital of Guizhou Medical University, Guizhou, 550004, P. R. China
| | - Wen Zhong
- Department of Pharmacy, Clinical Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Hospital of Guizhou Medical University, Guizhou, 550004, P. R. China
| | - Feier Xian
- West China School of Medicine, Sichuan University, Chengdu, 610040, P. R. China
| | - Mei Gao
- West China School of Medicine, Sichuan University, Chengdu, 610040, P. R. China
| | - Min Zeng
- West China School of Medicine, Sichuan University, Chengdu, 610040, P. R. China
| | - Jiehao Chen
- Animal Experiment Center, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, 610040, P. R. China
| | - Xiaoting Chen
- Animal Experiment Center, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, 610040, P. R. China
| | - Ming Li
- Department of Pharmacy, Clinical Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Hospital of Guizhou Medical University, Guizhou, 550004, P. R. China
| | - Kai Zhou
- Department of Orthopedics, West China Hospital, Sichuan University, 37# Wuhou Guoxue Road, Chengdu, 610040, P. R. China
| |
Collapse
|
17
|
Wang Z, Zheng X, Lin J, Zhou B, Zeng Z, Gao H, Chen H, Tang C. Electroacupuncture ameliorates cartilage damage in a rat model of knee osteoarthritis and regulates expression of miRNAs and the TLR4/NF-κB pathway. Acupunct Med 2025:9645284251342259. [PMID: 40390302 DOI: 10.1177/09645284251342259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
BACKGROUND Electroacupuncture (EA) has been shown to be effective in the treatment of knee osteoarthritis (KOA); however, its underlying mechanism remains unclear. METHODS 40 KOA model rats were divided into control, untreated model, EA-treated model and celecoxib-treated model groups (n=10 each). Articular cartilage of the knee joint was stained with hematoxylin and eosin (HE), periodic acid-Schiff (PAS) and Alcian blue (AB)-PAS, and Moran/Mankin scores were used to evaluate articular cartilage injury across groups. Moreover, toll-like receptor (TLR)4/nuclear factor (NF)-κB pathway (TN-P)-related protein levels in the articular cartilage were detected using Western blotting. Oxidative stress and inflammatory biomarkers in the synovial fluid were measured by enzyme-linked immunosorbent assay (ELISA). MicroRNA (miRNA/miR) expression was measured by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS Compared with the control group, Moran scores increased and Mankin scores decreased in the KOA model rats. In addition, compared with those in the control group, levels of superoxide dismutase (SOD), glutathione peroxidase (GSHPx) and interleukin (IL)-10 were significantly decreased, while levels of IL-1β, IL-6, tumor necrosis factor (TNF)-α, malondialdehyde (MDA) and nitric oxide (NO) were significantly increased, in the synovial fluid of the KOA model group. Protein levels of TLR4, anti-myeloid differentiation primary response protein 88 (MyD88) and p65 NF-κB phosphorylation were significantly increased in the articular cartilage of the KOA model group. EA and celecoxib treatment reversed the trends of these protein levels. Moreover, expression of miR-15a/127/140/146a/216a-5p and miR-27a-3p in the articular cartilage were markedly increased in KOA rats, while EA and celecoxib treatment reduced their expression. CONCLUSIONS EA reduces inflammation, oxidative stress and cartilage damage in KOA model rats, likely through regulation of the miRNA/TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Zhenzhen Wang
- Department of Rehabilitation, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiahai Zheng
- Department of Rehabilitation, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Lin
- Department of Rehabilitation, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bin Zhou
- Department of Rehabilitation, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenming Zeng
- Department of Rehabilitation, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiwei Gao
- Department of Rehabilitation, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haoxiong Chen
- Department of Rehabilitation, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunzhi Tang
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
18
|
Novy TCT, Joni IM, Lesmana R, Biben V, Setiawan. Chitosan Nanoparticles as an Alternative Therapeutic Approach for Knee Osteoarthritis Treatment: A Systematic Review. Int J Nanomedicine 2025; 20:6187-6203. [PMID: 40400782 PMCID: PMC12094476 DOI: 10.2147/ijn.s503829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/31/2025] [Indexed: 05/23/2025] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by the progressive breakdown of cartilage, leading to pain, inflammation, and reduced joint function. There are many variations of conventional therapies that exist, however, none proven to halt or reverse cartilage degradation. Chitosan, a biocompatible and biodegradable polysaccharide, has emerged as a promising candidate in OA treatment due to its chondroprotective properties, and ability to enhance chondrocyte proliferation and suppress inflammatory mediators. Recent advancements in nanotechnology have led to the development of chitosan nanoparticles (NPs), which offer a novel and effective approach for addressing the limitations associated with standard chitosan formulations, such as poor solubility and limited tissue penetration. Chitosan NPs have demonstrated superior bioavailability, sustained drug release, and targeted delivery, leading to improved therapeutic outcomes in preclinical models. This review explores evidence-based the therapeutic potential of chitosan NPs in the management of knee osteoarthritis, focusing on their role in cartilage regeneration and drug delivery.
Collapse
Affiliation(s)
| | - I Made Joni
- Functional Nano Powder University Center of Excellence (Finder U-Coe), Universitas Padjadjaran, Bandung, Indonesia
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung, Indonesia
| | - Ronny Lesmana
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Vitriana Biben
- Department of Physical Medicine and Rehabilitation, Dr. Hasan Sadikin General Hospital Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Setiawan
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
19
|
Tang W, Yin JB, Lin RG, Wu CY, Huang JL, Zhu JJ, Yang LF, Li GM, Cai DZ, Liu LL, Liu YL, Zhang HY. Rapgef3 modulates macrophage reprogramming and exacerbates synovitis and osteoarthritis under excessive mechanical loading. iScience 2025; 28:112131. [PMID: 40276767 PMCID: PMC12018577 DOI: 10.1016/j.isci.2025.112131] [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: 01/31/2024] [Revised: 04/11/2024] [Accepted: 02/26/2025] [Indexed: 04/26/2025] Open
Abstract
Evidence indicates that mechanical loading plays an important role in osteoarthritis (OA) progression, while the specific pathological changes of the synovium under excessive mechanical loading are unclear. Results showed that excessive mechanical loading caused pro-inflammation of synovial macrophages, which has been confirmed to exist in OA. High Rapgef3 expression level was found in RNA sequencing of RAW246.7 subjected to 0.5 Hz and 20% cyclic tensile strain. We verified this in the synovium of patients with OA and destabilization of the medial meniscus (DMM)-OA mice. Interestingly, the Rapgef3 content of chondrocytes was very low. Primary chondrocytes treated with Rapgef3 alone did not show metabolic phenotype, but an OA phenotype appeared when treated with Rapgef3-stimulated macrophage culture supernatant. Mechanically, excessive mechanical loading activated p65-nuclear factor κB (NF-κB) pathway through Rapgef3, which promoted the inflammation of macrophage, resulting in severe articular cartilage injury. Intra-articular Rapgef3 knockout reversed synovitis and cartilage degeneration, which might provide a therapeutic target for OA.
Collapse
Affiliation(s)
- Wen Tang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Jian-bin Yin
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Ren-gui Lin
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Chun-yu Wu
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Jia-luo Huang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Jin-jian Zhu
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Ling-feng Yang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Guang-ming Li
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Dao-zhang Cai
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Liang-liang Liu
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Yan-li Liu
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Hai-yan Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| |
Collapse
|
20
|
Battistoni CM, Munoz Briones J, Brubaker DK, Panitch A, Liu JC. Chondrogenic and chondroprotective response of composite collagen I/II-hyaluronic acid scaffolds within an inflammatory osteoarthritic environment. Biomater Sci 2025. [PMID: 40354044 DOI: 10.1039/d5bm00033e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Inflammation plays a key role in cartilage damage that occurs in osteoarthritis (OA). However, in vitro assessments of tissue-engineered constructs for cartilage regeneration generally do not consider their performance in the presence of inflammation. In this work, the chondrogenic differentiation potential of mesenchymal stromal cells (MSCs) was evaluated in the presence of both chondrogenic factors and inflammatory cytokines, and cartilage formation, degradative response, and inflammatory response were characterized. The addition of cytokines reduced cartilage production, increased cell proliferation, and resulted in an increase in inflammatory markers. Incorporation of hyaluronic acid (HA) had little impact on both collagen fibril microstructure and mechanical properties, two gel properties known to affect cell response, and thus allows the work to probe the biological impact of HA without the confounding effect of these gel properties. Regardless of in vitro environment, HA did not change cartilage production. The inflammatory response was similar with or without HA in terms of IL-6 and IL-10 secretion whereas IL-8 production exhibited some correlation with HA concentration as observed via a linear regression model. Additionally, in the presence of cytokines, inclusion of HA statistically decreased the gene- and protein-level expression of matrix metalloproteinase-13 (MMP-13). Thus, when exposed to both chondrogenic growth factors and inflammatory cytokines within a chondrogenic-promoting collagen I/II blended hydrogel, chondrogenic differentiation of MSCs was limited by the inflammatory environment. These findings emphasize the importance of understanding how biomaterials affect cell responses within disease-relevant inflammatory environments.
Collapse
Affiliation(s)
- Carly M Battistoni
- Davidson School of Chemical Engineering, Purdue, University, West Lafayette, IN 47907, USA.
| | - Javier Munoz Briones
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Purdue Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN 47907, USA
| | - Douglas K Brubaker
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH 44016, USA
- Blood Heart Lung Immunology Research Center, University Hospitals, Cleveland, OH 44106, USA
| | - Alyssa Panitch
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | - Julie C Liu
- Davidson School of Chemical Engineering, Purdue, University, West Lafayette, IN 47907, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
21
|
Cavallo M, Badley EM, Perruccio AV. Insomnia symptoms among individuals with osteoarthritis and symptoms indicative of osteoarthritis: A population-based cross-sectional study using the CLSA. PLoS One 2025; 20:e0322361. [PMID: 40354478 PMCID: PMC12068708 DOI: 10.1371/journal.pone.0322361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/20/2025] [Indexed: 05/14/2025] Open
Abstract
OBJECTIVE To compare the prevalence and examine the likelihood of insomnia symptoms in middle- and older-aged adults with an osteoarthritis (OA) diagnosis and with joint symptoms indicative of OA but without a diagnosis, in a population-based sample. METHODS Data are from the Canadian Longitudinal Study on Aging (participants aged >45). Individuals reported on doctor-diagnosed OA (hand, hip, or knee) and joint symptoms typical of OA, irrespective of OA diagnosis. A three-level 'OA-status' variable was derived: diagnosed OA; joint symptoms-no OA; no symptoms-no OA (controls). Participants responded to sleep-related questions and were categorized as experiencing insomnia symptoms (yes/no). Logistic regression analysis examined the association between insomnia symptoms and OA status, adjusting for a number of covariates. RESULTS Of 21,422 respondents, OA was reported by 29.1% (mean age 68.2) and 17.2% reported joint symptoms-no OA (mean age 63.9). One third of those with diagnosed OA and with joint symptoms-no OA reported insomnia symptoms compared to a quarter of controls. Those with OA and with joint symptoms-no OA were similarly more likely to report insomnia symptoms than controls (odds ratio (OR) 1.25, 95% CI 1.17-1.35 and OR 1.32, 95% CI 1.21-1.43). Also significantly associated with insomnia symptoms were female sex, current smoker, lower activity level, multiple chronic conditions and depressive symptoms. Odds ratio magnitudes were greatest for depressive symptoms (OR 2.50, 95% CI 2.26-2.76), comorbidity count (3 + vs. 0 OR 1.68, 95% CI 1.47-1.91) and female sex (OR 1.46, 95% 1.37-1.55). CONCLUSION Insomnia symptoms were not uncommon among comparatively younger individuals with typical OA joint symptoms and those with OA. This suggests that healthcare providers should address sleep-related issues in those consulting for joint pain, irrespective of diagnosis. Given their high prevalence, this also has implications for sleep-related issues at a population level.
Collapse
Affiliation(s)
- Melissa Cavallo
- Arthritis Community Research and Epidemiology Unit and Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Elizabeth M. Badley
- Arthritis Community Research and Epidemiology Unit and Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Anthony V. Perruccio
- Arthritis Community Research and Epidemiology Unit and Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Joseph YD, Ladd AL, Bhutani N. Hand Osteoarthritis: Molecular Mechanisms, Randomized Controlled Trials, and the Future of Targeted Treatment. Int J Mol Sci 2025; 26:4537. [PMID: 40429679 DOI: 10.3390/ijms26104537] [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: 02/10/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Hand osteoarthritis (OA) is a prevalent and disabling condition, yet its pathogenesis remains less studied than OA in large weight-bearing joints. Emerging genetic, epigenetic, and microbiome research suggests that hand OA might be biologically distinct, involving joint-specific pathways not shared by knee or hip OA. This review integrates genome-wide association studies specific to hand OA, highlighting key molecular contributors such as inflammatory cytokines. These genetic insights, together with emerging data on epigenetic alterations and gut microbial dysbiosis, point to broader systemic and regulatory influences on hand OA onset and progression. We also assess pharmacologic interventions tested in randomized controlled trials that have attempted to target these pathways. While agents such as TNF and IL-6 inhibitors, hydroxychloroquine, and corticosteroids have shown limited success, emerging evidence supports the potential of methotrexate in synovitis-positive general hand OA, platelet-rich plasma in thumb carpometacarpal (CMC) OA, and prolotherapy in interphalangeal (IP) OA. These findings illustrate the persistent gap between mechanistic understanding and therapeutic success. Future work must prioritize multifactorial strategies for addressing pain and translational frameworks that link molecular mechanisms to treatment response. In summary, this review offers an update on hand OA and identifies key opportunities for more targeted and effective therapy.
Collapse
Affiliation(s)
- Yemisi D Joseph
- Stanford University School of Medicine, Stanford University, Palo Alto, CA 94305, USA
| | - Amy L Ladd
- Department of Orthopaedic Surgery, Stanford University, Redwood City, CA 94063, USA
| | - Nidhi Bhutani
- Department of Orthopaedic Surgery, Stanford University, Redwood City, CA 94063, USA
| |
Collapse
|
23
|
Canalis E, Guzzo R, Schilling L, Denker E. NOTCH2 disrupts the synovial fibroblast identity and the inflammatory response of epiphyseal chondrocytes. J Biol Chem 2025:110206. [PMID: 40345585 DOI: 10.1016/j.jbc.2025.110206] [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: 03/20/2025] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025] Open
Abstract
Notch signaling plays a fundamental role in the inflammatory response and has been linked to the pathogenesis of osteoarthritis in murine models of the disease and in humans. To address how Notch signaling modifies transcriptomes and cell populations, we examined the effects of NOTCH2 in chondrocytes from mice harboring a NOTCH2 gain-of-function mutation (Notch2tm1.1Ecan) and a conditional NOTCH2 gain-of-function model expressing the NOTCH2 intracellular domain (NICD2) from the Rosa26 locus (R26-NICD2 mice). Bulk RNA-Sequencing (RNA-Seq) of primary epiphyseal cells from both gain-of-function models established increased expression of pathways associated with the phagosome, genes linked to osteoclast activity in rheumatoid arthritis signaling and pulmonary fibrosis signaling. Expression of genes linked to collagen degradation was enhanced in Notch2tm1.1Ecan cells, while genes related to osteoarthritis pathways were increased in NICD2-expressing cells. Single cell (sc)RNA-Seq of cultured Notch2tm1.1Ecan cells revealed clusters of cells related to limb mesenchyme, chondrogenic cells and fibroblasts including articular synovial fibroblasts. Pseudotime trajectory revealed close associations among clusters in control cultures, but the cluster of articular/synovial fibroblasts was disrupted in cells from Notch2tm1.1Ecan mice. ScRNA-Seq showed similarities in the cluster distributions and pseudotime trajectories of NICD2-expressing and control cells, except for altered progression in a cluster of NICD2-expressing cells. In conclusion, NOTCH2 enhances the activity of pathways associated with inflammation in epiphyseal chondrocytes and disrupts the transcriptome profile of articular/synovial fibroblasts.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030; Department of Medicine, UConn Health, Farmington, CT 06030; UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030.
| | - Rosa Guzzo
- Department of Neuroscience, UConn Health, Farmington, CT 06030
| | - Lauren Schilling
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030
| | - Emily Denker
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030
| |
Collapse
|
24
|
Li P, Zhang C, Yin W, Tao M, Niu Z, Cui Y, Wu D, Gao F. From bone marrow mesenchymal stem cells to diseases: the crucial role of m 6A methylation in orthopedics. Stem Cell Res Ther 2025; 16:228. [PMID: 40329380 PMCID: PMC12057228 DOI: 10.1186/s13287-025-04364-9] [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: 08/17/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Elucidating the molecular mechanisms underlying orthopedic diseases is crucial for guiding therapeutic strategies and developing innovative interventions. N6-methyladenosine (m6A)-an epitranscriptomic modification-has emerged as a key regulator of cellular fate and tissue homeostasis. Specifically, m6A plays a pivotal role in several RNA biological processes such as precursor RNA splicing, 3'-end processing, nuclear export, translation, and stability. Recent advancements indicate that m6A methylation regulates stem cell proliferation and osteogenic differentiation by modulating various signaling pathways. Extensive research has shown that abnormalities in m6A methylation contribute significantly to the onset and progression of various orthopedic diseases such as osteoporosis (OP), osteoarthritis (OA), rheumatoid arthritis (RA), and bone tumors. This review aims to summarize the key proteases involved in m6A methylation and their functions. The detailed mechanisms by which m6A methylation regulates osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) through direct and indirect ways are also discussed, with a focus on specific molecular pathways. Finally, this review analyzes the roles and mechanisms of m6A modification in the development and progression of multiple orthopedic diseases, offering a comprehensive understanding of the pathophysiology of these conditions and proposing new directions and molecular targets for innovative treatment strategies.
Collapse
Affiliation(s)
- Peng Li
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Chu Zhang
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Wen Yin
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Mijia Tao
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Zhipeng Niu
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Yutao Cui
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China.
| | - Dankai Wu
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China.
| | - Feng Gao
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China.
| |
Collapse
|
25
|
Jung B, Bhatti FUR, Mummareddy H, Kim Y, Park SH, Cho H. Targeted nanosome delivery of TPCA-1 for modulating inflammation in a mouse model of post-traumatic osteoarthritis. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2025; 67:102827. [PMID: 40324641 DOI: 10.1016/j.nano.2025.102827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/15/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Inflammation plays a significant role in the pathogenesis of knee post-traumatic osteoarthritis (PTOA) characterized by damage to cartilage and surrounding tissues that results in loss of physiological function. This inflammation is mainly regulated by NF-κB pathway. The TPCA-1 can inhibit IκB kinase (IKK) β in NF-κB pathway. Here, we optimized the delivery of TPCA-1 to the damaged knee joint via targeted nanosomes and examined its effects in a mouse model of PTOA. PTOA was induced in mice through a modified cyclic mechanical loading method. Mice were divided into groups receiving vehicle, TPCA-1 solution, or TPCA-1-loaded nanosomes. A concentration of 100 μM TPCA-1 was used based on preliminary studies. Control groups included untreated and vehicle-treated animals. Treatment efficacy was assessed using in vivo imaging, serum biochemical assays, gene expression analysis of cartilage tissues, histopathology, and behavioral analysis. Mechanical loading induced significant knee joint damage in the model. TPCA-1 nanosomes notably attenuated the adverse effects of loading, outperforming both the vehicle and TPCA-1-solution in reducing inflammation. Notably, serum levels of total NO and LDH were significantly lower in the TPCA-1-nanosome group. Inflammation, as indicated by MMP13 and IL1β gene expression, was substantially reduced. Enhanced cartilage preservation and function were confirmed through IVIS imaging, histological assessments, and improved behavior metrics. The targeted delivery of TPCA-1 via nanosomes effectively inhibits the NF-κB pathway, leading to significant reductions in inflammation and cartilage damage in a PTOA mouse model. This strategy demonstrates potential as a therapeutic intervention for managing inflammation and preserving joint health in osteoarthritis.
Collapse
Affiliation(s)
- Bongsu Jung
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Republic of Korea; Department of Biomedical Device, Gachon University, Republic of Korea
| | - Fazal-Ur-Rehman Bhatti
- Dept. of Orthopaedic Surgery & Biomedical Eng. University of Tennessee Health Science Center, Memphis, TN, USA
| | - Harisankeerth Mummareddy
- Dept. of Orthopaedic Surgery & Biomedical Eng. University of Tennessee Health Science Center, Memphis, TN, USA
| | - Youngjoo Kim
- Department of Biomedical Device, Gachon University, Republic of Korea
| | - Sang-Hyug Park
- Department of Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea.
| | - Hongsik Cho
- Dept. of Orthopaedic Surgery & Biomedical Eng. University of Tennessee Health Science Center, Memphis, TN, USA; Campbell Clinic, Memphis, TN, USA; VA Medical Center, Memphis, TN, USA.
| |
Collapse
|
26
|
Song M, Kim WJ, Shim J, Song K. Latilactobacillus sakei LB-P12 Ameliorates Osteoarthritis by Reducing Cartilage Degradation and Inflammation via Regulation of NF-κB/HIF-2α Pathway. J Microbiol Biotechnol 2025; 35:e2504013. [PMID: 40329628 PMCID: PMC12089955 DOI: 10.4014/jmb.2504.04013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage degradation, inflammation, and pain. Recent studies highlight the gut-joint axis, suggesting that gut microbiota influences joint health by modulating systemic inflammation and immune responses. This study investigated the effects of Latilactobacillus sakei LB-P12 on cartilage degradation and joint inflammation in a monosodium iodoacetate induced rat model of OA. OA severity was assessed through histological analysis, weight-bearing and micro-computed tomography (Micro-CT). Serum Interleukin 6 (IL-6) and prostaglandin E2 (PGE2) levels, along with interleukin-1β (Il1b) and matrix metalloproteinase 13 (Mmp13) expression in knee tissue, were measured. Then, the effect of L. sakei LB-P12 on inflammatory responses in interleukin-1β pretreated chondrocytes has also been investigated. The L. sakei LB-P12 improved weight-bearing distribution and reduced cartilage damage based on histological scores. Micro-CT showed increased bone volume fraction and bone mineral density. Treatment reduced serum IL-6 and PGE2 levels and suppressed Il1b and Mmp13 expression in knee tissues. In vitro, L. sakei LB-P12 inhibited lipopolysaccharide induced pro-inflammatory cytokines and nitric oxide production in macrophages. It also downregulated the expression of Epas1, which encodes hypoxia-inducible factor-2α (HIF-2α), and Mmp13 in IL-1β stimulated chondrocytes. L. sakei LB-P12 shows potential as a dietary supplement for alleviating OA-related pain, cartilage degradation, and inflammation by suppressing the nuclear factor-κB (NF-κB)/ HIF-2α pathway.
Collapse
Affiliation(s)
- Mikyung Song
- R&D Center, LISCure Biosciences Inc., Seongnam 13488, Republic of Korea
| | - Won Jun Kim
- R&D Center, LISCure Biosciences Inc., Seongnam 13488, Republic of Korea
| | - Jaeseok Shim
- R&D Center, LISCure Biosciences Inc., Seongnam 13488, Republic of Korea
| | - Kyoungsub Song
- R&D Center, LISCure Biosciences Inc., Seongnam 13488, Republic of Korea
| |
Collapse
|
27
|
Li W, Liu Y, Wei M, Yang Z, Li Z, Guo Z, Yan L, Lu Y, Tang H, Li B, Huang W. Functionalized Biomimetic Nanoparticles Targeting the IL-10/IL-10Rα/Glycolytic Axis in Synovial Macrophages Alleviate Cartilage Degeneration in Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504768. [PMID: 40317692 DOI: 10.1002/advs.202504768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Indexed: 05/07/2025]
Abstract
Osteoarthritis (OA) is a low-grade inflammatory disease that is highly associated with severe hyperplasia of the synovial membrane and the degeneration of cartilage. Interleukin-10 (IL-10), has been extensively studied, while its receptor, IL-10Rα, has not been widely mentioned in the context of OA. A significant difference is found in the expression of IL-10Rα in synovial macrophages from normal and OA patients, along with a marked increase in the glycolytic activity of synovial macrophages. In IL-10RαLysm OA mice, the specific deficiency of IL-10Rα exacerbated the progression of OA. Mechanistically, hypoxia-inducible factor-1α (HIF-1α) is identified as a key transcription factor, and its inhibition significantly weakened the glycolytic process. Additionally, differences in ferroptosis of chondrocytes are observed. After co-culturing the two types of cells in vitro, a significant connection is found between the glycolytic state of synovial macrophages and the ferroptosis of chondrocytes. To achieve targeted therapy, MI@UN, a biomimetic nanoparticle encapsulating NO-prednisolone in UIO-66-NH2, surface-modified with IL-10, and coated with macrophage membranes (MM), is developed. It significantly slows osteoarthritis progression in mice. This offers new insights into OA pathogenesis, highlighting IL-10Rα as a therapeutic target and supporting MI@UN's translational use for OA treatment.
Collapse
Affiliation(s)
- Wenwei Li
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
| | - Yang Liu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Ming Wei
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Zhichao Yang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Zhaoyu Li
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Zezhong Guo
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
| | - Liang Yan
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yang Lu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Hao Tang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Bofeng Li
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, 230001, China
| | - Wei Huang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| |
Collapse
|
28
|
Hou W, Shang X, Hao X, Pan C, Zheng Z, Zhang Y, Deng X, Chi R, Liu J, Guo F, Sun K, Xu T. SHP2-mediated ROS activation induces chondrocyte paraptosis in osteoarthritis and is attenuated by low-intensity pulsed ultrasound. J Orthop Translat 2025; 52:233-248. [PMID: 40337549 PMCID: PMC12056802 DOI: 10.1016/j.jot.2025.04.005] [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: 10/31/2024] [Revised: 03/20/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025] Open
Abstract
Background Paraptosis is a novel form of programmed cell death, generally caused by disrupted proteostasis or alterations of redox homeostasis. However, its impact and underlying mechanisms on the pathology of osteoarthritis (OA) are still unclear. This study aimed to investigate the role and regulatory mechanism of SHP2 in chondrocyte paraptosis and the effects influenced by low-intensity pulsed ultrasound (LIPUS). Methods SHP2, a MAPK upstream intermediary, has been identified as one of the critical targets of IL-1β-induced paraptosis in the GEO and GeneCard databases. The expression of SHP2 in chondrocytes was regulated by either siRNA knockdown or plasmid overexpression. Additionally, adeno-associated viruses were injected into the knee joints of rats to explore whether SHP2 plays a role in the development of OA. The impact of LIPUS on paraptosis and OA was examined in IL-1β-induced chondrocytes and a post-traumatic OA model, with SHP2 regulation assessed at both cellular and animal levels. Results An increase in cellular reactive oxygen species (ROS) caused by IL-1β halts the growth of chondrocytes and induces paraptosis in the chondrocytes. IL-1β-induced paraptosis, manifested as endoplasmic reticulum (ER)-derived vacuolization, was mediated by ROS-mediated ER stress and MAPK activation. SHP2 facilitates ROS production, thereby exacerbating the chondrocytes paraptosis. SHP2 knockdown and ROS inhibition effectively reduced this process and significantly mitigated inflammation and cartilage degeneration. Furthermore, we discovered that LIPUS delayed OA progression by inhibiting the activation of the MAPK pathway, ER stress, and ER-derived vacuoles in chondrocytes, all of which play critical roles in paraptosis, through the downregulation of SHP2 expression. Results on animals showed that LIPUS inhibited cartilage degeneration and alleviated OA progression. Conclusion SHP2 exacerbates IL-1β-induced oxidative stress and the subsequent paraptosis in chondrocytes, promoting OA progression. LIPUS mitigates paraptosis by modulating SHP2, which in turn slows OA progression. The translational potential of this article This study indicates that a novel SHP2-mediated cell death mechanism, paraptosis, plays a role in post-traumatic OA progression. LIPUS helps maintain cartilage-subchondral bone unit integrity by targeting SHP2 inhibition. SHP2 emerges as a potential therapeutic target, while LIPUS provides a promising non-invasive approach for treating trauma-related OA.
Collapse
Affiliation(s)
- Wenjie Hou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xingru Shang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Rehabilitation Medicine,Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chunran Pan
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zehang Zheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiwen Zhang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaofeng Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ruimin Chi
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
29
|
Yang W, Hu L, Tian Y, Yang Y, Guo W, Xiao K, Yang R, Yang H, Zhou Z, Cheng C. WTAP improves chondrocyte loss and dysfunctions to ameliorate osteoarthritis through mediating the mA methylation and mRNA stability of IL-33. Int J Biol Macromol 2025; 306:141330. [PMID: 39984079 DOI: 10.1016/j.ijbiomac.2025.141330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/13/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Osteoarthritis (OA) is a multifactorial degenerative disorder entailing cartilage loss and progressive joint failure. m6A RNA methylation could impact multiple disorders, including OA. In this study, m6A methylation regulator WTAP was down-regulated in OA cartilage, accompanied by significantly lower m6A methylation levels in OA tissues. In the DMM-induced mice OA model and IL-1β- or TNF-α-stimulated chondrocytes, WTAP and m6A methylation levels were decreased, but IL-1β, IL-6, and TNF-α cytokine expressions were elevated. In vivo and in vitro, WTAP overexpression increased m6A methylation levels but reduced proinflammatory cytokine contents. Furthermore, WTAP overexpression (OE) increased chondrocyte viability and proliferation, aggrecan and collagen II protein, and decreased cell apoptosis, MMP3, MMP13, and ADAMTS5. WTAP-mediated m6A methylation of IL-33 and impaired IL-33 mRNA stability. IL-33 OE caused no changes to WTAP expression; however, IL-33 OE partially attenuated WTAP OE-induced IL-33 downregulation. IL-33 overexpression inhibited chondrocyte viability and proliferation, decreased aggrecan and collagen II but elevated MMP3, MMP13, and ADAMTS5, and increased cell apoptosis and proinflammatory cytokine contents. More importantly, IL-33 eliminated the effects of WTAP OE on chondrocytes. Therefore, WTAP is down-regulated in OA; WTAP improves chondrocyte proliferation and function, thereby ameliorating OA through mediating m6A methylation of IL-33 and impairing IL-33 mRNA stability.
Collapse
Affiliation(s)
- Wenjian Yang
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China
| | - Lianghua Hu
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China
| | - Ye Tian
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China
| | - Yufan Yang
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China
| | - Wei Guo
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China
| | - Kai Xiao
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China
| | - Ruiqi Yang
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China
| | - Hua Yang
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China
| | - Zhihong Zhou
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China; Yiyang Medical College, Yiyang, Hunan 413000, China
| | - Chao Cheng
- Department of Orthopaedics, Yiyang Central Hospital, Hunan University of Chinese Medicine, Yiyang, Hunan 413000, China; Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, Hunan 413000, China; The fourth people's hospital of Yiyang city, Yiyang, Hunan 413000, China.
| |
Collapse
|
30
|
Li M, Deng T, Chen Q, Jiang S, Li H, Li J, You S, Xie HQ, Shen B. A versatile platform based on matrix metalloproteinase-sensitive peptides for novel diagnostic and therapeutic strategies in arthritis. Bioact Mater 2025; 47:100-120. [PMID: 39897588 PMCID: PMC11787566 DOI: 10.1016/j.bioactmat.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/11/2025] [Accepted: 01/12/2025] [Indexed: 02/04/2025] Open
Abstract
Matrix metalloproteinases (MMPs), coupled with other proteinases and glycanases, can degrade proteoglycans, collagens, and other extracellular matrix (ECM) components in inflammatory and non-inflammatory arthritis, making them important pathogenic molecules and ideal disease indicators and pharmaceutical intervention triggers. For MMP responsiveness, MMP-sensitive peptides (MSPs) are among the most easily synthesized and cost-effective substrates, with free terminal amine and/or carboxyl groups extensively employed in multiple designs. We hereby provide a comprehensive review over the mechanisms and advances in MSP applications for the management of arthritis. These applications include early and precise diagnosis of MMP activity via fluorescence probe technologies; acting as nanodrug carriers to enable on-demand drug release triggered by pathological microenvironments; and facilitating cartilage engineering through MMP-mediated degradation, which promotes cell migration, matrix synthesis, and tissue integration. Specifically, the ultra-sensitive MSP diagnostic probes could significantly advance the early diagnosis and detection of osteoarthritis (OA), while MSP-based drug carriers for rheumatoid arthritis (RA) can intelligently release anti-inflammatory drugs effectively during flare-ups, or even before symptoms manifest. The continuous progress in MSP development may acceleratedly lead to novel management regimens for arthropathy in the future.
Collapse
Affiliation(s)
- Mingyang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tao Deng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Quan Chen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shenghu Jiang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiayi Li
- Department of Nephrology, The People's Hospital of Yubei District of Chongqing, Chongqing, China
| | - Shenglan You
- Animal Imaging Core Facilities, West China Hospital, Sichuan University, China
| | - Hui-qi Xie
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Stem Cell and Tissue Engineering Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Shen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
31
|
Sun L, Wang Y, Kan T, Wang H, Cui J, Wang L, Liu C, Li H, Yu Z, Yan M. Elevated expression of Piezo1 activates the cGAS-STING pathway in chondrocytes by releasing mitochondrial DNA. Osteoarthritis Cartilage 2025; 33:601-615. [PMID: 39978573 DOI: 10.1016/j.joca.2025.02.778] [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: 09/19/2024] [Revised: 01/27/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVE Abnormal mechanical stress is a key factor in osteoarthritis (OA) pathogenesis. This study aims to investigate the role of the mechanosensitive ion channel Piezo1 in activating the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway and its contribution to cartilage degradation in OA. METHODS We conducted both in vivo and in vitro experiments. In vitro, chondrocytes were subjected to mechanical stress, and Piezo1 expression, calcium ion (Ca2+) influx, and mitochondrial permeability changes were analyzed. In vivo, Piezo1 conditional knockout (Col2a1CreERT; Piezo1flox/flox) mice were used to assess the activation of the cGAS-STING pathway and cartilage degradation. Additionally, the effects of STING inhibitors on inflammation and OA progression were evaluated. RESULTS Mechanical stress significantly increased Piezo1 expression and Ca2+ influx in chondrocytes, leading to mitochondrial Ca2+ overload and mitochondrial DNA (mtDNA) release. This triggered activation of the cGAS-STING pathway (9.35[95%Confidence Interval (CI) 1.378 to 18.032], n=3 biologically independent samples), resulting in inflammatory responses (4.185[95%CI 0.411 to 8.168], n=3 biologically independent samples). In Piezo1 knockout mice, cGAS-STING activation (-7.23[95%CI -10.52 to -3.89], n=6) and cartilage degradation (Osteoarthritis Research Society International (OARSI) grade; -3.651[95%CI -5.562 to -1.681] n=6) were reduced. STING inhibitors effectively decreased inflammation (-8.95[95%CI -17.24 to -1.31], n=3 biologically independent samples) and slowed OA progression (OARSI grade; -2.76 [95%CI -4.37 to -1.08], n=6) in both in vivo and in vitro models. CONCLUSIONS Mechanical stress induces mtDNA release via Piezo1 activation, which triggers the cGAS-STING pathway and exacerbates cartilage degradation. Targeting Piezo1 or the cGAS-STING pathway may offer a promising therapeutic strategy to reduce inflammation and protect cartilage in OA.
Collapse
Affiliation(s)
- Lin Sun
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyou Kan
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Han Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junqi Cui
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liao Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenglei Liu
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Hanjun Li
- Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Mengning Yan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
32
|
Zhou K, Yuan M, Sun J, Zhang F, Li X, Xiao X, Wu X. Co-delivery of IL-1Ra and SOX9 via AAV inhibits inflammation and promotes cartilage repair in surgically induced osteoarthritis animal models. Gene Ther 2025; 32:211-222. [PMID: 39833570 DOI: 10.1038/s41434-025-00515-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Osteoarthritis (OA), a prevalent joint disorder, can lead to disability, with no effective treatment available. Interleukin-1 (IL-1) plays a crucial role in the progression of OA, and its receptor antagonist (IL-1Ra), a natural IL-1 inhibitor, represents a promising therapeutic target by obstructing the IL-1 signaling pathway. This study delivered IL-1Ra via adeno-associated virus (AAV), a gene therapy vector enabling long-term protein expression, to treat knee osteoarthritis (KOA) in animal models. scAAV-oIL-1Ra-I1/2 injected directly into the joint in both MMT/ACLT-induced KOA model rat improved abnormal gait (increasing footprint area and pressure), subchondral bone lesions, and significantly reduced cartilage wear and pathological scores. In the MMT-induced KOA rabbit model, weight-bearing asymmetry (indicating pain) improved after 8 weeks of scAAV-oIL-1Ra-I1/2 administration, and X-ray showed decreased K-L scores (severity grade), reduced cartilage loss, and lower pathology scores compared to untreated animals. Additionally, sex-determining region Y-type high mobility group box 9 (SOX9) was co-delivered with IL-1Ra via AAV in ACLT + MMT-induced KOA rats. The combined treatment significantly alleviated subchondral bone lesions, cartilage destruction, synovial inflammation, and pathological scores, demonstrating superior efficacy compared to either treatment administered alone. Co-delivering IL-1Ra and SOX9 inhibited IL-1 mediated inflammatory signaling, maintained cartilage homeostasis, and promoted its repair in KOA models, suggesting potential for clinical use.
Collapse
Affiliation(s)
- Kaiyi Zhou
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Meng Yuan
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiabao Sun
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Feixu Zhang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Xinting Li
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Xiao Xiao
- School of Pharmacy, East China University of Science and Technology, Shanghai, China.
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China.
| | - Xia Wu
- School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
33
|
Lee SA, Lee SB, Kim DK, Lee SY, Kim CS. Pyropia yezoensis Extract Attenuates Osteoarthritis Progression In Vitro and In Vivo. Prev Nutr Food Sci 2025; 30:141-151. [PMID: 40352296 PMCID: PMC12061538 DOI: 10.3746/pnf.2025.30.2.141] [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: 12/24/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 05/14/2025] Open
Abstract
Osteoarthritis (OA), a degenerative disease characterized by cartilage degradation and inflammation, occurs due to trauma caused by external stimuli or cartilage aging. Pyropia yezoensis is a red alga that belongs to the Porphyra family and is consumed as food in Asia, especially Korea, Japan, and China. P. yezoensis contains various bioactive substances, including carotenoids, flavonoids, and vitamins, that exert anti-inflammatory, antioxidant, and anti-photoaging effects. In the present study, the anti-osteoarthritic effects of 30% fermented alcohol extract of P. yezoensis (30% FEPY) on interleukin-1 beta (IL-1β)-stimulated chondrocytes and a destabilization of the medial meniscus (DMM)-induced OA rat model were investigated. The results showed that pretreatment with 30% FEPY significantly reduced the IL-1β-induced expression of inflammatory factors (e.g., inducible nitric oxide synthase and cyclooxygenase-2) and cartilage-degrading enzymes [matrix metalloproteinase (MMP) 1, MMP3, MMP13, a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) 4, and ADAMTS5], which was analyzed using Griess reaction, enzyme-linked immunosorbent assay, and Western blot analysis. The anti-osteoarthritic effects of 30% FEPY, which were mediated through mitogen-activated protein kinase and nuclear factor kappa-light-chain-enhancer of activated B cell signaling, were analyzed using Western blot analysis. In an in vivo study, Safranin O staining and immunohistochemistry analysis revealed that treatment with 30% FEPY significantly increased cartilage degradation and collagen type II protein expression in the DMM group. These findings collectively suggest that 30% FEPY is a promising candidate for alleviating OA progression and developing new therapeutic drugs.
Collapse
Affiliation(s)
- Seul Ah Lee
- Department of Oral Biochemistry, College of Dentistry, Chosun University, Gwangju 61452, Korea
| | - Seul Bi Lee
- Marine Healthcare Research & Evaluation Center, Chosun University, Jeonnam 59146, Korea
| | - Do Kyung Kim
- Oral Biology Research Institute, College of Dentistry, Chosun University, Gwangju 61452, Korea
| | - Sook-Young Lee
- Marine Healthcare Research & Evaluation Center, Chosun University, Jeonnam 59146, Korea
| | - Chun Sung Kim
- Department of Oral Biochemistry, College of Dentistry, Chosun University, Gwangju 61452, Korea
| |
Collapse
|
34
|
Sun Y, Luo Z, Fu Y, Ngo T, Wang W, Wang Y, Kong Y. Primary cilia and inflammatory response: unveiling new mechanisms in osteoarthritis progression. Exp Biol Med (Maywood) 2025; 250:10490. [PMID: 40357414 PMCID: PMC12066368 DOI: 10.3389/ebm.2025.10490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that can lead to chronic pain and disability. The pathogenesis of OA involves chronic low-grade inflammation, characterized by the degradation of chondrocytes, inflammation of the synovium, and systemic low-grade inflammation. This inflammatory response accelerates the progression of OA and contributes to pain and functional impairment. Primary cilia play a crucial role in cellular signal transduction and the maintenance of cartilage matrix homeostasis, and their dysfunction is closely linked to inflammatory responses. Given these roles, primary cilia may significantly contribute to the pathogenesis of OA. This review explores inflammation-associated signaling pathways in OA, including NF-κB, MAPK, JAK/STAT, and PI3K/AKT/mTOR signaling. In addition, we place particular emphasis on cilia-mediated inflammatory modulation in OA. Primary cilia mediate chondrocyte responses to mechanical loading and inflammatory cytokines via pathways including NF-κB, MAPK, TRPV4, and Hedgehog signaling. Notably, alterations in the length and incidence of primary cilia in chondrocytes during OA further underscore their potential role in disease pathogenesis. The identification of biomarkers and therapeutic targets related to primary cilia and inflammatory pathways offers new potential for the treatment and management of OA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ying Kong
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
35
|
Shao C, Chen H, Liu T, Pan C. The Hippo pathway in bone and cartilage: implications for development and disease. PeerJ 2025; 13:e19334. [PMID: 40292098 PMCID: PMC12024444 DOI: 10.7717/peerj.19334] [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: 11/13/2024] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Bone is the main structure of the human body; it mainly plays a supporting role and participates in metabolic processes. The Hippo signaling pathway is composed of a series of protein kinases, including the mammalian STE20-like kinase MST1/2 and the large tumor suppressor LATS1/2, which are widely involved in pathophysiological processes, including cell proliferation, differentiation, apoptosis and death, especially those related to biomechanical transduction in vivo. However, the role of it in regulating skeletal system development and the evolution of bone-related diseases remains poorly understood. The pathway can intervene in and regulate the physiological activities of bone-related cells such as osteoclasts and chondrocytes through its own or other bone-related signaling pathways, such as the Wnt pathway, the Notch pathway, and receptor activator of nuclear factor-κB ligand (RANKL), thereby affecting the occurrence and development of bone diseases. This article discusses the role of the Hippo signaling pathway in bone development and disease to provide new insights into the treatment of bone-related diseases by targeting the Hippo signaling pathway.
Collapse
Affiliation(s)
- Chenwei Shao
- Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Hao Chen
- Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Tingting Liu
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Chun Pan
- Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
36
|
Knothe Tate ML. The Flow of Life: Convergent Approaches to Understanding Musculoskeletal Health from Molecular- to Meso-Length Scales. FRONT BIOSCI-LANDMRK 2025; 30:25231. [PMID: 40302317 DOI: 10.31083/fbl25231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 05/02/2025]
Abstract
In the current perspective and review article, we address the human body as a living ecosystem with collecting watersheds and draining hydrosheds; we integrate our discoveries over the past quarter of a century and pose the critical open research questions to be addressed going forward, with the aim to improve cell, tissue, organ and organismal health. First, we address the flow of fluid through the tissues of the musculoskeletal system, after which we describe the interactions of the fluid, at multiple lengths and time scales, with the molecular to macroscopic non-fluid tissue components, discussing bone and tissues in the context of "living" chromatography and/or electrophoresis columns. Thereafter, we discuss the implications of functional barrier integrity, and the effects of cytokines on active barrier function and molecular transport between organ systems, tissue compartments, and within tissues. In addition, we address the fluid and its flow and the multi-physics implications thereof for the living inhabitants of tissues, i.e., the cells. Finally, we describe the implications of the solid and fluid components and the cellular inhabitants on ecosystem health, where the tissues and organs comprise the organism form interacting ecosystems throughout life and in the context of health and disease. By taking convergent approaches to understanding musculoskeletal, human and environmental health (which themselves are interdependent), we hope to pave new paths of innovation and discovery, to improve the lives of our worlds' inhabitants, from the worlds of our bone and joints and bodies to the interacting ecosystems of our Earth to unknown worlds beyond our current understanding.
Collapse
Affiliation(s)
- Melissa Louise Knothe Tate
- Blue Mountains World Interdisciplinary Innovation Institute, Blue Mountains National Park, NSW 2782, Australia
| |
Collapse
|
37
|
Li S, Wang S, Zhang L, Ka Y, Zhou M, Wang Y, Tang Z, Zhang J, Wang W, Liu W. Research progress on pharmacokinetics, anti-inflammatory and immunomodulatory effects of kaempferol. Int Immunopharmacol 2025; 152:114387. [PMID: 40054326 DOI: 10.1016/j.intimp.2025.114387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025]
Abstract
Chronic inflammation (an abnormal state) and autoimmune disease (AD) can both cause multiple organ damage. AD is a heterogeneous group of diseases due to immune dysfunction. Chronic inflammation is closely related to AD and is an important part of AD. With the increasing prevalence of AD, researchers are constantly exploring new drugs with small side effects, considerable curative effects, and lower costs. Kaempferol, a flavonoid, possesses a range of biological functions, including antioxidant, anti-inflammatory, anti-neoplastic, and immunomodulatory capabilities. This compound is prevalent in a variety of plant sources, such as vegetables, fruits, and medicinal herbs traditionally used in Chinese medicine. A plethora of empirical evidence from animal-based research supports the assertion that this particular substance exhibits both anti-inflammatory and immunomodulatory effects, with the curative effect being significant and application prospects. This article mainly summarizes and discusses the pharmacokinetics, drug delivery system, and the mechanism of kaempferol on immune cells, cytokines, signaling pathways, and other aspects. This paper summarizes the existing kaempferol drug delivery system, analyzes the possibility and limitations of kaempferol as a new anti-inflammatory and immunomodulatory drug, and discusses how to apply it in clinical practice. Therefore, kaempferol can more effectively exert its anti-inflammatory and immune-modulating effects, thereby demonstrating therapeutic potential in clinical settings, while reducing patient burden.
Collapse
Affiliation(s)
- Suiran Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Siwei Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Lei Zhang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, China
| | - Yuxiu Ka
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Meijiao Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yiwen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Zhuo Tang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Jiamin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
38
|
Wang Y, Gu M, Zheng Z, Jiang H, Han L, Huang H, Wu Y, Li C. Therapeutic approaches for SAPHO syndrome from the perspective of pathogenesis: a review of the literature. Front Immunol 2025; 16:1560398. [PMID: 40303415 PMCID: PMC12037609 DOI: 10.3389/fimmu.2025.1560398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/19/2025] [Indexed: 05/02/2025] Open
Abstract
Synovitis, acne, pustulosis, hyperostosis and osteitis (SAPHO) syndrome is a rare autoinflammatory disease characterized by cutaneous manifestations and osteoarticular damage. The pathogenesis of SAPHO syndrome has not yet been elucidated, but studies have shown that the abnormal bone metabolism of patients with SAPHO syndrome is most likely due to localized infections that induce immune disorders in the body. Although no standardized treatment protocols exist, based on existing case studies and data from open studies, we propose that the treatment of SAPHO syndrome can be categorized into three areas according to the symptomatic manifestations of the disease: (1) control of focal infections using antibiotics and tonsillectomy; (2) administration of DMARDs to manage disease progression; and (3) bone remodeling therapy with bisphosphonates to address abnormal bone metabolism. Furthermore, a comprehensive treatment approach tailored to the clinical manifestations of the patient can effectively alleviate symptoms and enhance quality of life.
Collapse
Affiliation(s)
- Yunuo Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Mengjiao Gu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zixiang Zheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Haixu Jiang
- School of Chinese Materia, Beijing University of Chinese Medicine, Beijing, China
| | - Luyao Han
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Hanjing Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuanhao Wu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chen Li
- Department of Dermatology, Tianjin Institute of Integrative Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
- Department of Rheumatology, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
39
|
Heijman MWJ, van den Ende CHM, Cornel JH, Smolders JMH, Schers HJ, Kievit W, Koeter S, van den Bemt BJF, Popa CD. Design of a randomised, placebo-controlled, double-blind multicentre study assessing the effect of colchicine on the incidence of knee or hip replacements in symptomatic knee or hip osteoarthritis: the ECHO trial. BMJ Open 2025; 15:e098096. [PMID: 40228852 PMCID: PMC11997832 DOI: 10.1136/bmjopen-2024-098096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/21/2025] [Indexed: 04/16/2025] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is a multifactorial disease in which low-grade inflammation is considered to play a pivotal role. Although colchicine is a widely used anti-inflammatory drug in the treatment of gout, its effect in OA is still disputed due to inconsistent results of short-term clinical trials. Therefore, we aim to evaluate the effect of long-term colchicine 0.5 mg once daily on the incidence of knee or hip replacements in patients with knee or hip OA. METHODS AND ANALYSIS The ECHO trial is a prospective, multicentre, randomised, double-blind, placebo-controlled, phase III trial in which 1200 participants with knee or hip OA tolerant to colchicine during a 30-day run-in period will be 1:1 randomised to colchicine 0.5 mg once daily or matching placebo using concealed allocation. The primary endpoint is the time from randomisation to the first knee or hip replacement assessed up to 4.5 years. Secondary endpoints include course of pain, physical function, joint space narrowing, low-grade inflammation, quality of life, clinical or radiological onset of OA in a new joint group other than present at baseline, number of participants using pain medication during the study, onset of new cardiovascular events (ie, myocardial infarction, ischaemia-driven coronary revascularisation, ischaemic stroke, peripheral artery disease or cardiovascular death) and direct and indirect costs related to treatment and disease burden due to OA. Harm-related endpoints include the number of (serious) adverse events, the number of withdrawals due to (serious) adverse events and changes in laboratory data (ie, serum creatinine, estimated glomerular filtration rate and alanine transferase) throughout the study. The primary analysis will be performed according to the intention-to-treat principle. ETHICS AND DISSEMINATION This trial has been approved by the Medical Ethics Review Committee East-Netherlands. Findings will be presented at scientific meetings and published in a peer-reviewed scientific journal. TRIAL REGISTRATION NUMBER NCT06578182.
Collapse
Affiliation(s)
- Michelle W J Heijman
- Department of Research, Sint Maartenskliniek, Nijmegen, Netherlands
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cornelia H M van den Ende
- Department of Research, Sint Maartenskliniek, Nijmegen, Netherlands
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jan H Cornel
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, Netherlands
- Dutch Network for Cardiovascular Research (WCN), Utrecht, Netherlands
| | - José M H Smolders
- Department of Orthopedics, Sint Maartenskliniek, Nijmegen, Netherlands
| | - Henk J Schers
- Department of Primary and Community Care, Radboud University Medical Center, Nijmegen, Netherlands
| | - Wietske Kievit
- Department of Health Evidence, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sander Koeter
- Department of Orthopedics, Canisius Wilhelmina Hospital, Nijmegen, Netherlands
| | - Bart J F van den Bemt
- Department of Pharmacy, Sint Maartenskliniek, Nijmegen, Netherlands
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - Calin D Popa
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Rheumatology, Sint Maartenskliniek, Nijmegen, Netherlands
| |
Collapse
|
40
|
Wang SQ, Peng Z, Sun H, Han YM, Zhang B, Pineda L, Boerboom G, Sun LH, Liu Y, Deng ZC. Evaluating the Impact of an Organic Trace Mineral mix on the Redox Homeostasis, Immunity, and Performance of Sows and their Offspring. Biol Trace Elem Res 2025; 203:1798-1807. [PMID: 38980512 DOI: 10.1007/s12011-024-04300-7] [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: 05/17/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
The objective of the study was to evaluate the effects of trace mineral supplementation in sows during gestation and lactation on the performance and health status of sows and their offspring. Sows (n = 30; Landrace × Yorkshire; avg parity = 3.9) were randomly allocated into two dietary treatments. Sows received a basal diet supplemented with 12 mg/kg Cu, 30 mg/kg Fe, 90 mg/kg Zn, 70 mg/kg Mn, 0.30 mg/kg Se, and 1.5 mg/kg I from an inorganic trace mineral source (ITM) or a blend of hydroxychloride and organic trace mineral source (HOTM) from day 1 of gestation until the end of the lactation period at day 21. Compared to the ITM, the HOTM supplementation increased (P < 0.05) both litter birth weight and individual piglet birth weight. Although not statistically significant, HOTM tended to increase (P = 0.069) the level of lactose in colostrum. HOTM increased (P < 0.05) the concentration of Mn and Se in the colostrum, milk, and serum of sows and/or piglets. Notably, the Zn concentration in the serum of sows was higher in sows supplemented with ITM compared to HOTM. Moreover, HOTM increased (P < 0.05) the activities of GPX and SOD in gestating sows and piglets, as well as increased (P < 0.05) cytokines (IL-1β, TNF-α, and IL-10) in the serum of sows. The immunoglobulins (IgA, IgG, and IgM) also increased in sows and/or piglets at certain experimental time points. In conclusion, HOTM supplementation positively affected piglet development and improved the health status of sows and piglets potentially by regulating redox homeostasis and immunity.
Collapse
Affiliation(s)
- Shao-Qing Wang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Zhe Peng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Hua Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Inner Mongolia Academy of Agriculture and Animal Husbandry Science, Hohhot, 010031, Inner Mongolia, China
| | - Yan-Ming Han
- Selko Feed Additives, Nutreco, Amersfoort, The Netherlands
| | - Bo Zhang
- Selko Feed Additives, Nutreco, Amersfoort, The Netherlands
| | - Lane Pineda
- Selko Feed Additives, Nutreco, Amersfoort, The Netherlands
| | - Gavin Boerboom
- Selko Feed Additives, Nutreco, Amersfoort, The Netherlands
| | - Lv-Hui Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
| | - Ying Liu
- Tianjin Animal Disease Prevention and Control Center, Tianjin, 300402, China.
| | - Zhang-Chao Deng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
| |
Collapse
|
41
|
Joukar A, Karnik S, Noori-Dokht H, Younesi S, Trippel SB, Wagner DR. Mechanical Wear of Degraded Articular Cartilage. Ann Biomed Eng 2025; 53:956-965. [PMID: 39863807 DOI: 10.1007/s10439-025-03680-8] [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: 09/26/2024] [Accepted: 01/12/2025] [Indexed: 01/27/2025]
Abstract
PURPOSE To evaluate the mechanical wear of cartilage with different types of degradation. METHODS Bovine osteochondral explants were treated with interleukin-1β (IL-1β) to mimic inflammatory conditions, with chondroitinase ABC (ChABC) to specifically remove glycosaminoglycans (GAGs), or with collagenase to degrade the collagen network during 5 days of culture. Viscoelastic properties of cartilage were characterized via indentation. Biochemical assays were performed to quantify the cartilage matrix loss to the media during culture and from an accelerated, ex vivo wear test. The coefficient of friction during the wear test was measured. Distribution of GAGs in the tissue was assessed histologically. RESULTS All three degradative treatments decreased the cartilage modulus values and depleted GAGs in histological sections. However, wear was not uniform among the different treatments. Collagen loss from the tissue due to mechanical wear was only higher with IL-1β and collagenase treatment, while collagen loss due to wear with ChABC treatment was similar to untreated controls. In addition, less GAG was released due to mechanical wear in all degraded groups than the controls, likely because GAGs had already been depleted from these tissues during culture. As no significant differences in the coefficient of friction were observed between groups, changes in wear were attributed to altered tissue composition and structure rather than to changes in frictional forces. CONCLUSIONS Results suggest that cartilage with a degraded collagen network is more susceptible to mechanical wear, but that cartilage wear may be relatively unaffected by the loss of GAGs. Furthermore, exacerbated mechanical wear could be an additional mechanism by which inflammatory cytokines induce cartilage breakdown.
Collapse
Affiliation(s)
- Amin Joukar
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Sonali Karnik
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 723 W. Michigan Street, SL 260, Indianapolis, IN, 46202, USA
| | - Hessam Noori-Dokht
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Sogol Younesi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Stephen B Trippel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 723 W. Michigan Street, SL 260, Indianapolis, IN, 46202, USA
| | - Diane R Wagner
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 723 W. Michigan Street, SL 260, Indianapolis, IN, 46202, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
42
|
Peng Y, Demidchik V, Li Y, Shen Z. Comparison of terpenoids in Nauclea officinalis and Paederia scandens and their anti-inflammatory effects on RAW264.7 macrophages. Fitoterapia 2025; 182:106411. [PMID: 39909359 DOI: 10.1016/j.fitote.2025.106411] [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: 10/18/2024] [Revised: 01/14/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Terpenoids are important components that exert pharmacological effects in Rubiaceae plants. In this study, two Rubiaceae plants, Nauclea officinalis and Paederia scandens, which are widely distributed in Hainan, China, were collected. The extracts of these two plants were obtained through boiling water extraction and analyzed using UHPLC-ESI-QE-Orbitrap-MS. By comparing with the mzCloud database, terpenoids with a matching rate of 85 % were identified. The results revealed that the aqueous extracts of N. officinalis mainly contain six pentacyclic triterpenoids, one diterpenoid, and one iridoid. The aqueous extracts of P. scandens contains one monoterpenoid, one diterpenoid, two pentacyclic triterpenes, and 4 iridoids. To verify the anti-inflammatory efficacy of the two extracts, in this study, they were added to the lipopolysaccharide (LPS)-induced RAW264.7 macrophages inflammation model. The results showed that the two extracts can reduce the secretion of proinflammatory cytokines IL-1β, IL-6, and TNF-α (p < 0.05) and the mortality rate of cell inflammation (p < 0.05) by inhibiting the activation of the NF-κB/NLRP3 pathway (p < 0.05). The study identified the terpenoids in N. officinalis and P. scandens and verified their anti-inflammatory effects in the RAW264.7 macrophages inflammation model.
Collapse
Affiliation(s)
- Yuxuan Peng
- Hainan College of Vocation and Technique, 95 Nanhai Ave., 570100 Haikou, China; Department of Plant Cell Biology and Bioengineering, Biology Faculty, Belarusian State University, 4 Independence Ave., 220030 Minsk, Belarus.
| | - Vadim Demidchik
- Institute of Experimental Botany, National Academy of Sciences of Belarus, 27 Botanichskaya St., 220072 Minsk, Belarus; International Research Centre for Environmental Membrane Biology and Department of Horticulture, Foshan University, Foshan, China
| | - Yan Li
- International Sakharov Environmental Institute, Belarusian State University, 15 Botanichskaya St., 220030 Minsk, Belarus
| | - Zhenguo Shen
- Hainan College of Vocation and Technique, 95 Nanhai Ave., 570100 Haikou, China.
| |
Collapse
|
43
|
Xu Y, Yang Y, Song H, Li M, Shi W, Yu T, Lin J, Yu Y. The Role of Exerkines in the Treatment of Knee Osteoarthritis: From Mechanisms to Exercise Strategies. Orthop Surg 2025; 17:1021-1035. [PMID: 39854050 PMCID: PMC11962297 DOI: 10.1111/os.14365] [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: 09/04/2024] [Revised: 12/25/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
With the increasing prevalence of knee osteoarthritis (KOA), the limitations of traditional treatments, such as their limited efficacy in halting disease progression and their potential side effects, are becoming more evident. This situation has prompted scientists to seek more effective strategies. In recent years, exercise therapy has gained prominence in KOA treatment due to its safety, efficacy, and cost-effectiveness, which are underpinned by the molecular actions of exerkines. Unlike conventional therapies, exerkines offer specific advantages by targeting inflammatory responses, enhancing chondrocyte proliferation, and slowing cartilage degradation at the molecular level. This review explores the potential mechanisms involved in and application prospects of exerkines in KOA treatment and provides a comprehensive analysis of their role. Studies show that appropriate exercise not only promotes overall health, but also positively impacts KOA by stimulating exerkine production. The effectiveness of exerkines, however, is influenced by exercise modality, intensity, and duration of exercise, making the development of personalized exercise plans crucial for KOA patients. Based on these insights, this paper proposes targeted exercise strategies designed to maximize exerkine benefits, aiming to provide novel perspectives for KOA prevention and treatment.
Collapse
Affiliation(s)
- Yuxiong Xu
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Yizhuo Yang
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Hanan Song
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Ming Li
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Weihao Shi
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Tongwu Yu
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Jianhao Lin
- Arthritis Clinic & Research CenterPeking University People's HospitalBeijingChina
| | - Yanli Yu
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| |
Collapse
|
44
|
Chen Y, Xu N, Zhang W, Wang Y, Su T, Zhou Y, Xu J. FSH enhances the inflammatory response of macrophages in the knee joint possibly through the NFκB pathway. FEBS Open Bio 2025; 15:622-633. [PMID: 39801258 PMCID: PMC11961395 DOI: 10.1002/2211-5463.13959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 10/22/2024] [Accepted: 12/13/2024] [Indexed: 04/03/2025] Open
Abstract
Previous studies have suggested that women with higher follicle-stimulating hormone (FSH) levels have a greater incidence of osteoarthritis (OA) compared to women with lower FSH despite normal estrogen levels. Our previous studies also showed that FSH has a negative effect on cartilage in postmenopausal OA. However, no studies have investigated the effect of FSH on the synovium. Here, we showed that the FSH receptor (FSHR) is expressed on RAW264.7 cells and BMDM (Bone Marrow-Derived Macrophages), and found that FSH stimulation promotes the production and secretion of inflammatory cytokines in synovial macrophages. In RAW264.7 cells, FSH stimulation enhances phosphorylation and nuclear translocation of P65, suggesting the activation of NFκB signaling, while the knockdown of FSHR eliminates the proinflammatory effect of FSH. To further validate these results, we used an ovariectomy mouse model supplemented with FSH and estrogen, and a mouse model with FSH neutralization. We noted that FSHR was expressed on mouse synovial joint membranes. Furthermore, in ovariectomy mice supplemented with estrogen and treated with FSH, synovial macrophages were significantly increased, while the opposite was the case in the FSH neutralizing group, which suggest that FSH triggers an inflammatory response in the synovial tissue in mice. Taken together, our results indicate that FSH is an important regulator in synovial inflammation via NFκB signaling activation and, to some extent, appears to accelerate the development of osteoarthritis.
Collapse
Affiliation(s)
- Yu Chen
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Na Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Wen‐wen Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Yan Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Tong Su
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| | - Yan‐man Zhou
- Department of NephrologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Jin Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain AgingMinistry of EducationJinanChina
- Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanChina
- “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic DiseasesJinanChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanChina
| |
Collapse
|
45
|
Perrone G, Giampaoli C, Smirnoff AL, Ochoa A, Pareja R, De Simone E. Early effects of anti-TNFα antibodies in horses with osteoarthritis. J Equine Vet Sci 2025; 147:105384. [PMID: 39956345 DOI: 10.1016/j.jevs.2025.105384] [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: 04/09/2024] [Revised: 12/20/2024] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Osteoarthritis (OA) is the most common pathology in sport horses, presenting a chronic and recurrent course. Traditional anti-inflammatory treatments provide only temporary clinical improvement and pain relief; however, they have been associated with metabolic disorders in articular cartilage in the medium term. In this study, we generated a llama polyclonal antibody against equine TNF-α, which was used for intra-articular treatment in horses with OA. Treatment effects were assessed through clinical examination and the determination of inflammatory parameters (MMP-2, MMP-9, LDH, and GAGs) in synovial fluid prior to treatment (T0) and on day 30 (T30). Treated animals showed significant improvement in the clinical score, which decreased from 8.47 ± 2.57 to 6.16 ± 1.71 (mean ± SD) on T30 (p < 0.001). Additionally, a reduction in the activity of MMP-2 (188 % ± 82.99 to 147 % ± 40.6 %, p < 0.05), MMP-9 (100 % ± 61.28 to 74.37 % ± 64.26, p < 0.05), and LDH (239.3 IU/l ± 147.4 to 143 IU/l ± 61.21, p < 0.01) was observed. Furthermore, on T30, GAGs levels were significantly increased (1.167 ± 0.46 mg/ml to 1.439 ± 0.267 mg/ml, p < 0.01). These results indicate that the local blockade of pro-inflammatory molecules such as TNF-α reduces pain and the levels of inflammatory molecules associated with catabolic processes and joint damage.
Collapse
Affiliation(s)
- G Perrone
- Chair of Equine Health and Production, Faculty of Veterinary Sciences, University of Buenos Aires, Av. Chorroarín 280 (C1427CWO), Buenos Aires, Argentina
| | - C Giampaoli
- Chair of Animal Physiology, Faculty of Veterinary Sciences, University of Buenos Aires, Av. Chorroarín 280 (C1427CWO), Buenos Aires, Argentina
| | - A Lorenzo Smirnoff
- Chair of Animal Physiology, Faculty of Veterinary Sciences, University of Buenos Aires, Av. Chorroarín 280 (C1427CWO), Buenos Aires, Argentina
| | - A Ochoa
- Chair of Animal Physiology, Faculty of Veterinary Sciences, University of Buenos Aires, Av. Chorroarín 280 (C1427CWO), Buenos Aires, Argentina
| | - R Pareja
- Chair of Animal Physiology, Faculty of Veterinary Sciences, University of Buenos Aires, Av. Chorroarín 280 (C1427CWO), Buenos Aires, Argentina
| | - E De Simone
- Chair of Animal Physiology, Faculty of Veterinary Sciences, University of Buenos Aires, Av. Chorroarín 280 (C1427CWO), Buenos Aires, Argentina.
| |
Collapse
|
46
|
Wang Y, Chen X, Chen Y, Sun Q, Wang H. Regulatory effect and mechanism of CircSEC24A in IL-1β-induced osteoarthritis. Arch Physiol Biochem 2025; 131:188-198. [PMID: 39328069 DOI: 10.1080/13813455.2024.2404975] [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: 04/02/2024] [Revised: 06/27/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by articular cartilage degeneration and damage. Increasing circular RNAs (circRNAs) have been identified to participate in the pathogenesis of OA. Hsa_circ_0128006 (also known as circSEC24) was reported as an upregulated circRNA in OA tissues, but its biological role and underlying mechanism in OA are still to be discussed. circSEC24A and NAMPT expression levels were upregulated, and miR-515-5p was reduced in OA cartilage tissues and IL-1β-treated CHON-001 cells. The absence of circSEC24A overturned IL-1β-induced suppression of cell viability and promotion of oxidative stress, apoptosis, extracellular matrix (ECM) degradation, and inflammation in CHON-001 cells. Mechanistically, circSEC24A acted as a molecular sponge for miR-515-5p to affect NAMPT expression. CircSEC24A knockdown could attenuate IL-1β-triggered CHON-001 cell injury partly via the miR-515-5p/NAMPT axis, providing new insight into the underlying application of circSEC24A in OA treatment.
Collapse
Affiliation(s)
- Yuanrui Wang
- Department of Orthopaedics, Xijing Hospital, The Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Xiaochao Chen
- Department of Orthopaedics, Xijing Hospital, The Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Yongfeng Chen
- Department of Orthopaedics, Xijing Hospital, The Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Qiang Sun
- Department of Orthopaedics, Xijing Hospital, The Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Huayi Wang
- Department of Orthopaedics, Xijing Hospital, The Air Force Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
47
|
Moulin D, Sellam J, Berenbaum F, Guicheux J, Boutet MA. The role of the immune system in osteoarthritis: mechanisms, challenges and future directions. Nat Rev Rheumatol 2025; 21:221-236. [PMID: 40082724 DOI: 10.1038/s41584-025-01223-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 03/16/2025]
Abstract
Osteoarthritis (OA) is a chronic joint disease that has long been considered a simple wear-and-tear condition. Over the past decade, research has revealed that various inflammatory features of OA, such as low-grade peripheral inflammation and synovitis, contribute substantially to the pathophysiology of the disease. Technological advances in the past 5 years have revealed a large diversity of innate and adaptive immune cells in the joints, particularly in the synovium and infrapatellar fat pad. Notably, the presence of synovial lymphoid structures, circulating autoantibodies and alterations in memory T cell and B cell populations have been documented in OA. These data indicate a potential contribution of self-reactivity to the disease pathogenesis, blurring the often narrow and inaccurate line between chronic inflammatory and autoimmune diseases. The diverse immune changes associated with OA pathogenesis can vary across disease phenotypes, and a better characterization of their underlying molecular endotypes will be key to stratifying patients, designing novel therapeutic approaches and ultimately ameliorating treatment allocation. Furthermore, examining both articular and systemic alterations, including changes in the gut-joint axis and microbial dysbiosis, could open up novel avenues for OA management.
Collapse
Affiliation(s)
- David Moulin
- Université de Lorraine, CNRS, IMoPA, Nancy, France.
- CHRU-Nancy, IHU INFINY, Nancy, France.
| | - Jérémie Sellam
- Department of Rheumatology, Saint-Antoine Hospital, Centre de Recherche Saint-Antoine, Inserm, Sorbonne Université UMRS 938, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Francis Berenbaum
- Department of Rheumatology, Saint-Antoine Hospital, Centre de Recherche Saint-Antoine, Inserm, Sorbonne Université UMRS 938, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, INSERM, CHU Nantes, UMR1229 Regenerative Medicine and Skeleton, RMeS, Nantes, France
| | - Marie-Astrid Boutet
- Nantes Université, Oniris, INSERM, CHU Nantes, UMR1229 Regenerative Medicine and Skeleton, RMeS, Nantes, France.
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, UK.
| |
Collapse
|
48
|
Fang X, Zhang H, Zhou H, Shen S, Lao Z, Zhang Z, Bian Y, Zhou C, Jin H, Tong P, Huang Y, Zhou H, Zeng H, Fu F, Wu C, Zheng W, Ruan H. Systemic Lupus Erythematosus Exacerbates Hip Arthritis by Promoting Chondrocyte Pyroptosis in the Femoral Head via Activating the NF-κB Pathway. J Cell Mol Med 2025; 29:e70531. [PMID: 40179133 PMCID: PMC11967699 DOI: 10.1111/jcmm.70531] [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: 01/03/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterised by chronic inflammation and immune dysregulation, significantly impacting multiple organ systems, including the joints. While SLE is known to contribute to musculoskeletal complications, its role in hip arthritis development and the underlying mechanisms remain poorly understood. This study aims to investigate the relationship between SLE and hip arthritis progression using MRL/lpr mice, which exhibit early-onset SLE, compared with MRL/MpJ control mice at 14 weeks of age. Through comprehensive histological, immunohistochemical and molecular analyses, we evaluated articular cartilage (AC) degeneration, extracellular matrix (ECM) metabolism, inflammatory responses, and chondrocyte pyroptosis. Our results demonstrated that MRL/lpr mice developed an accelerated hip arthritis-like phenotype, manifesting as enhanced AC degeneration, impaired chondrocyte proliferation, heightened apoptosis and promoted inflammatory cytokine production. Notably, SLE markedly stimulated chondrocyte pyroptosis by increasing pyroptosis-related proteins, including NLRP3, ASC, CASPASE-1 and GSDMD, via activating the NF-κB pathway. These findings establish a novel mechanistic link between SLE and hip arthritis progression, demonstrating that SLE promotes chondrocyte pyroptosis to exacerbate AC degeneration via NF-κB activation, highlighting chondrocyte pyroptosis as a key driver of SLE-associated hip arthritis and a potential therapeutic target for mitigating SLE-induced joint manifestations.
Collapse
Affiliation(s)
- Xuliang Fang
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Helou Zhang
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Huiqing Zhou
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Shuchao Shen
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Zhaobai Lao
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Zhiguo Zhang
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Yishan Bian
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Chengcong Zhou
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Hongting Jin
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Peijian Tong
- Department of OrthopaedicsThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Yanqun Huang
- Hangzhou Fuyang Hospital of TCM Orthopedics and TraumatologyHangzhouChina
| | - Hong Zhou
- Hangzhou Fuyang Hospital of TCM Orthopedics and TraumatologyHangzhouChina
| | - Hanbing Zeng
- The Second Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Fangda Fu
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Chengliang Wu
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Wenbiao Zheng
- Department of OrthopedicsTaizhou Municipal HospitalTaizhouChina
| | - Hongfeng Ruan
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| |
Collapse
|
49
|
Li J, Sun F, Zhang Y, Pan X, Li B, Zhang G, Zhou Q. MiR-103-3p regulates chondrocyte autophagy, apoptosis, and ECM degradation through the PI3K/Akt/mTOR pathway by targeting CPEB3. J Orthop Surg Res 2025; 20:324. [PMID: 40155964 PMCID: PMC11954267 DOI: 10.1186/s13018-025-05719-x] [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: 01/16/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Chondrocyte apoptosis is associated with the severity of cartilage destruction and matrix degeneration in the progression of osteoarthritis. Increasing evidence indicates that autophagy has a significant cytoprotective effect against chondrocyte apoptosis. Here, we investigated the role of microRNA-103-3p (miR-103-3p) in regulating chondrocyte function and elucidated the underlying mechanism. METHODS MiR-103-3p expression in interleukin-1β (IL-1β)-stimulated chondrocytes was evaluated using RT-qPCR. The targets of miR-103-3p predicted by online databases were verified using biotin-based pulldown assay and luciferase reporter assay. IL-1β stimulated-chondrocytes were transfected with miR-103-3p inhibitor along with siRNA targeting cytoplasmic polyadenylation element-binding protein3 (siCPEB3), the autophagy inhibitor 3-MA, or the PI3K agonist 740 Y-P. Chondrocyte proliferation was evaluated using cell counting kit-8. Apoptosis was detected by flow cytometry. The levels of apoptosis-, extracellular matrix (ECM)-, autophagy-, and the PI3K/Akt/mTOR pathway-related proteins in chondrocytes were detected using immunoblotting or immunofluorescence. RESULTS We found that IL-1β stimulation upregulated miR-103-3p and downregulated CPEB3 in mouse chondrocytes. Inhibiting miR-103-3p reduced IL-1β-induced apoptosis and ECM macromolecule degradation while enhancing autophagy in chondrocytes. MiR-103-3p targeted CPEB3, and its downregulation rescued the expression of level in IL-1β stimulated-chondrocytes. MiR-103-3p downregulation inhibited the PI3K/Akt/mTOR pathway in IL-1β stimulated-chondrocytes by upregulating CPEB3. 3-MA, 740 Y-P, or CPEB3 knockdown counteracted the effect of miR-103-3p downregulation on chondrocyte apoptosis, ECM macromolecule degradation, and autophagy. CONCLUSION Overall, inhibition of miR-103-3p reduces IL-1β-induced apoptosis and ECM macromolecule degradation in chondrocytes by enhancing autophagy through the CPEB3/PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Jun Li
- Department of Orthopedics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Hungshi, 435000, China
| | - Farui Sun
- Department of Orthopedics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Hungshi, 435000, China
| | - Yuanjin Zhang
- Department of Orthopedics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Hungshi, 435000, China
| | - Xian Pan
- Department of Orthopedics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Hungshi, 435000, China
| | - Bo Li
- Department of Orthopedics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Hungshi, 435000, China
| | - Guofu Zhang
- Department of Orthopedics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Hungshi, 435000, China
| | - Qian Zhou
- Department of Geriatrics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Tianjin Avenue No. 141, Huangshigang District, 435000, Hungshi, Hubei Province, China.
| |
Collapse
|
50
|
Lai P, Ma Y, Sang W, Zhou Q, Chen H, Wang C, Yin J, Wang T, Zhu L, Zhou X, He C, Ma J. Reprogramming Macrophage Phenotype Using a Reactive Oxygen Species-Responsive Liposome Delivery System for Inflammation Microenvironment Remodeling and Osteoarthritis Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:17932-17947. [PMID: 40094857 DOI: 10.1021/acsami.4c19160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The progression of osteoarthritis (OA) is closely linked to synovial inflammation caused by an imbalance between M1 and M2 macrophages. To tackle this problem, we developed a liposome responsive to reactive oxygen species (ROS), modified with folic acid ligands to target M1-polarized macrophages, and loaded with the anti-inflammatory agent dimethyl fumarate (DMF). This liposome-based drug delivery system was designed to reprogram macrophage phenotype to remodel the inflammatory microenvironment in the joint cavity and alleviate OA degeneration. The liposome we prepared had a suitable size and negative zeta potential, with uniform size, good stability in aqueous solution, and excellent biocompatibility. Laboratory tests showed that these DMF-filled liposomes notably decreased high levels of ROS in M1-type macrophages and shifted macrophage polarization via the Nrf2/HO-1 pathway, which in turn lessened inflammation in chondrocytes and averted their apoptosis. Additionally, animal studies demonstrated that liposomes containing DMF exhibited notable anti-inflammatory properties, significantly reduced synovial inflammation, safeguarded injured cartilage, reversed changes in subchondral bone, and effectively slowed the progression of osteoarthritis in a mouse model induced by anterior cruciate ligament transection (ACLT). Therefore, ROS-responsive liposomes targeting M1-polarized macrophages represent a promising and valuable approach for OA treatment.
Collapse
Affiliation(s)
- Peng Lai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yichao Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Weilin Sang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Qiang Zhou
- Department of Otolaryngology, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital), Wenzhou 325200, China
| | - Hongjie Chen
- The School of Medicine, Nankai University, Tianjin 300071, China
| | - Cong Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jian Yin
- Department of Orthopedics, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing 211100, China
| | - Tao Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Libo Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiaojun Zhou
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Chuanglong He
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|