1
|
Wu YJ, Li SM, Chen CL, Chen ZR, Chen JJ. Anti-inflammatory activity of Pogostemon cablin: Bioactive components and their modulation of MAPK and NF-κB signaling pathway. Bioorg Chem 2025; 161:108516. [PMID: 40345124 DOI: 10.1016/j.bioorg.2025.108516] [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: 02/12/2025] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025]
Abstract
Pogostemon cablin is a well-known Lamiaceae plant and widely utilized in Traditional Chinese Medicine (TCM) for its neuroprotective, anti-inflammatory, and anxiolytic properties. In this study, 16 known compounds (1-15 and 17) and one semi-synthesized new compound, 5-hydroxy-3-isoprenyloxy-7,3',4'-trimethoxyflavone (16), including flavonoids, pyranones, sesquiterpenes, and benzenoids, were obtained and characterized from aerial parts of P. cablin and investigated for their anti-inflammatory properties through the MAPK and NF-κB signaling pathways in LPS-induced RAW264.7 macrophages. Among the isolated compounds, rhamnazin (4), pachypodol (5), and (E)-2-methyl-6-(p-tolyl)hept-3-en-2-ol (15) exhibited potent anti-inflammatory activities in LPS-induced RAW264.7 macrophages. Rhamnazin (4) significantly modulated IκBα levels and reduced the expressions of phosphorylation of JNK and p38, indicating its effects on suppressing NF-κB activation and mitigating inflammation via MAPK signaling. Pachypodol (5) selectively inhibited iNOS and p-JNK expressions, showing specificity in its anti-inflammatory activity. (E)-2-Methyl-6-(p-tolyl)hept-3-en-2-ol (15) downregulated iNOS, p-Erk, and p-JNK expressions, demonstrating a broader inhibitory profile on pro-inflammatory mediators. Further molecular docking results demonstrated bioactive compounds 4, 5, and 15 possessed strong binding affinities with key residues, particularly Hem901, Pro344, and Glu371, consistent with their NO inhibition effects. In addition, in silico prediction of physicochemical properties confirmed favorable oral bioavailability and drug-likeness, supporting their potential as lead compounds for anti-inflammatory drug development. These findings provide comprehensive molecular insight into the anti-inflammatory effects and reveal the therapeutic potential of P. cablin constituents as natural plant-derived NF-κB and MAPK-targeting anti-inflammatory agents, offering promising candidates for managing inflammatory diseases.
Collapse
Affiliation(s)
- Yu-Jing Wu
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Sin-Min Li
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Zih-Rong Chen
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Jih-Jung Chen
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404333, Taiwan; Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan.
| |
Collapse
|
2
|
Hou J, Li Y, Zhang Y, Yang N, Chen B, Ma G, Zhu N. Integrated network pharmacology reveals the mechanism of action of Xianlinggubao prescription for inflammation in osteoarthritis. BMC Complement Med Ther 2025; 25:190. [PMID: 40426157 PMCID: PMC12108044 DOI: 10.1186/s12906-025-04928-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA), a leading cause of disability worldwide, is characterized by complex interactions between cartilage degradation and synovial inflammation. While NSAIDs are the primary treatment, their prolonged use exacerbates gastrointestinal risks and does not alter disease progression. Xianlinggubao (XLGB), an approved Chinese herbal remedy for osteoporosis, has demonstrated promising anti-osteoarthritic effects in preliminary studies. However, its multi-component mechanisms targeting OA-related inflammation require further clarification. This study integrates network pharmacology with experimental validation to investigate XLGB's anti-inflammatory mechanisms in OA. METHODS Bioactive compounds of XLGB and their respective targets were sourced from the TCMSP, ETCM, SymMap, and ChEMBL databases. Targets linked to OA-related inflammation were identified through differential expression analysis and by querying OMIM, GeneCards, and PubMed Gene databases. Network pharmacology and bioinformatics approaches were employed to construct compound-target and protein-protein interaction (PPI) networks, enabling the identification of pivotal therapeutic targets. Functional enrichment of these targets was performed using the ClusterProfiler package in R. The binding affinity of compounds to anti-inflammatory OA targets was assessed through molecular docking, dynamics simulations, RT-PCR, and immunofluorescence assays. RESULTS Fifty-five bioactive compounds corresponding to 475 XLGB targets and 125 genes involved in OA-related inflammation were identified. PPI network analysis revealed that XLGB may alleviate OA inflammation by modulating key genes, including COX-2, IL-1β, TNF, IL-6, and MMP-9. Molecular simulations indicated strong binding affinities between bioactive compounds in XLGB and these critical targets. Functional enrichment analysis suggested that XLGB's anti-inflammatory action in OA may involve regulation of pathways such as IL-17, TNF, and NF-κB. In vitro experiments further confirmed that XLGB mitigates OA inflammation by modulating these genes, proteins, and signaling pathways. CONCLUSIONS Through network pharmacology, this study elucidated the mechanisms of XLGB in OA inflammation, highlighting its modulation of IL-6, IL-1β, TNF-α, PTGS2, MMP-9, and the NF-κB pathway. These findings provide strong support for the clinical application of XLGB in managing OA-related inflammation.
Collapse
Affiliation(s)
- Jingyi Hou
- Hebei Province Key Laboratory of Study and Exploitation of Chinese Medicine, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei, China
| | - Yubo Li
- Department of Minimally Invasive Spinal Surgery, The Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi Street, Chengde, Hebei, 067000, China
| | - Yu Zhang
- Department of Minimally Invasive Spinal Surgery, The Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi Street, Chengde, Hebei, 067000, China
| | - Ning Yang
- Department of Minimally Invasive Spinal Surgery, The Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi Street, Chengde, Hebei, 067000, China
| | - Bin Chen
- Department of Minimally Invasive Spinal Surgery, The Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi Street, Chengde, Hebei, 067000, China
| | - Guiyun Ma
- Department of Minimally Invasive Spinal Surgery, The Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi Street, Chengde, Hebei, 067000, China.
| | - Naiqiang Zhu
- Department of Minimally Invasive Spinal Surgery, The Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi Street, Chengde, Hebei, 067000, China.
- Hebei Key Laboratory of Panvascular Diseases, Chengde, Hebei, China.
| |
Collapse
|
3
|
Jia L, Yuan J, Chen Y, Liang P, Wu J, Xie Y, Bai X, Wang Y, Geng W. Lactobacillus kefiranofaciens and its enhancement effect on the anti-inflammatory function of CII/Gln in MIA-induced osteoarthritis by protecting the intestinal barrier and gut microecology. Int J Food Sci Nutr 2025:1-14. [PMID: 40401730 DOI: 10.1080/09637486.2025.2508173] [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: 01/21/2025] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Osteoarthritis (OA) is a globally chronic disease affecting middle-aged and elderly individuals, with growing evidences implicating gut microbiota in its pathogenesis. Lactobacillus kefiranofaciens ZW3 (ZW3) shows potential in modulating gut microbiota, protecting intestinal barrier, and regulating immunity. This study explored the therapeutic effects of ZW3, alone and combined with CII/Gln, using a monoiodoacetate (MIA)-induced OA model. Results indicated that ZW3 significantly mitigated cartilage damage and inflammation alone or combined with CII/Gln, possibly by improving intestinal integrity, reduced oxidative stress, and regulated MMPs expression. 16S rDNA sequencing showed ZW3, especially with CII/Gln, increased beneficial bacteria of Ruminococcaceae and Lachnospiraceae abundances. Furthermore, ZW3 or with CII/Gln elevated SCFAs levels in intestinal contents in OA rats. These findings propose a novel probiotic-based strategy, potentially combined with functional foods, for OA intervention and treatment.
Collapse
Affiliation(s)
- Longgang Jia
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Jiahu Yuan
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Yu Chen
- Binhai New Area Hospital of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Peixin Liang
- Tangshan Normal University, Tangshan, Hebei, P. R. China
| | - Jiangtao Wu
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Yufeng Xie
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
- College of Food Science and Engineering, Harbin University, Harbin, P. R. China
| | - Xiaojia Bai
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Yanping Wang
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Weitao Geng
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| |
Collapse
|
4
|
He L, Gong X, Guo H, Zhou K, Lan Y, Lv M, Liu X, Lin S, Hua Y, Guo J, Fan Z, Li Y. Single cell RNA-sequencing identified CCR7+/RELB+/IRF1+ T cell responding for juvenile idiopathic arthritis pathogenesis. Front Immunol 2025; 16:1528446. [PMID: 40406113 PMCID: PMC12095314 DOI: 10.3389/fimmu.2025.1528446] [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: 11/14/2024] [Accepted: 04/21/2025] [Indexed: 05/26/2025] Open
Abstract
Background To further explore the disease heterogeneity of different subtypes of Juvenile idiopathic arthritis (JIA) and analyze their pathogenesis mechanisms. Method The single-cell RNA sequencing (scRNA-seq) analysis of peripheral blood mononuclear cells (PBMCs) was carried out to investigate the disease heterogeneity and molecular mechanisms of immune responses in immune cells in JIA. Result In our study, we provided a immunological landscape of HLA-B27-positive JIA and HLA-B27-negative JIA immune cells at single cell RNA-Seq resolution. We found a higher proportion of CCR7+/RELB+/IRF1+ triple positive T cells in the peripheral blood of patients with JIA, and such T cells were predominantly present in HLA-B27+ JIA patients. Furthermore, we hypothesized that CCR7+/RELB+/IRF1+ triple positive T cells were highly activated T cells capable of promoting the differentiation of osteoclasts by producing IL-17, thus causing damage to cartilage in HLA-B27+ JIA patients. Unlike JIA patients, CCR7+/RELB+/IRF1+ triple positive T cells were not found in the peripheral blood of pSS patients and SLE patients, moreover, T cells from pSS patients and SLE patients were less able to produce IL-17 than those from JIA patients. Conclusion Our study provided evidence of cellular and molecular levels of involvement in JIA pathogenesis and identified the critical roles for T cells in JIA pathogenesis. Furthermore, our results suggested that there were significant differences in T cell composition and gene expression between HLA-B27+ JIA patients and HLA-B27- JIA patients. Our findings indicated that CCR7+/RELB+/IRF1+ positive T cells could damage the cartilage of HLA-B27+ JIA by producing cytokines such as IL-17.
Collapse
Affiliation(s)
- Lewei He
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xue Gong
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Guo
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaiyu Zhou
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yue Lan
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingyi Lv
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoliang Liu
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sha Lin
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yimin Hua
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, China
| | - Zhenxin Fan
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yifei Li
- Key Laboratory of Bioresources and Eco-Environment of MOE, College of Life Sciences, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Yuan YS, Li HY, Lu H, Li GC, Cao Z, Xu C, Xiao HH, Zhang LP, Xu HL. Reprogramming mitochondrial metabolism to enhance macrophages polarization by ROS-responsive nanoparticles for osteoarthritis. Biomaterials 2025; 322:123395. [PMID: 40403559 DOI: 10.1016/j.biomaterials.2025.123395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025]
Abstract
Osteoarthritis (OA) is a chronic low-grade inflammatory joint disease closely related to the inflammatory pathological microenvironment caused by synovial M1 macrophages. In contrast to the proinflammatory role of M1 macrophages, M2 macrophages contribute to anti-inflammatory responses and tissue repair. Therefore, shifting the M2/M1 phenotype ratio in favor of M2 macrophages has become a promising therapeutic strategy for OA. However, current therapeutics cannot penetrate the synovium and only show limited drug retention time. Herein, we developed an OA microenvironment-responsive nanocarrier with thioketal bonds in the main chain and β-1,3-d-glucan and triphenylphosphine units in the side chain, which can respond to reactive oxygen species (ROS) and target macrophages and mitochondrial aggregation. For OA treatment, 4-octyl itaconate and dexamethasone were encapsulated within the nanocarrier, forming HBPTG@OD that effectively eliminated mitochondrial ROS and inducible nitric oxide synthase in M1 macrophages. HBPTG@OD significantly suppressed the release of inflammatory factors by macrophages and thereby reducing chondrocyte death. In vivo studies in a destabilized medial meniscus (DMM)-induced OA model showed that HBPTG@OD effectively converted M1 synovial macrophages to M2 macrophages, consequently delaying chondrogenic apoptosis. This study presents a nanocarrier-based strategy that effectively repolarizes M1 macrophages, demonstrating great promise for the treatment of OA.
Collapse
Affiliation(s)
- Yu-Song Yuan
- Department of Trauma and Orthopedics, Foot and Ankle Center, Peking University People's Hospital, 11th Xizhimen South Street, Beijing, China, 100044; Department of Trauma and Orthopedics, China-Japan Friendship Hospital, 2nd Yinghuayuan East Street, Beijing, China, 100029
| | - Hui-Yun Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China; Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Lu
- Department of Trauma and Orthopedics, Foot and Ankle Center, Peking University People's Hospital, 11th Xizhimen South Street, Beijing, China, 100044
| | - Gui-Cheng Li
- Department of Trauma and Orthopedics, Foot and Ankle Center, Peking University People's Hospital, 11th Xizhimen South Street, Beijing, China, 100044
| | - Zheng Cao
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, 4006, Australia
| | - Hai-Hua Xiao
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China.
| | - Ling-Pu Zhang
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China.
| | - Hai-Lin Xu
- Department of Trauma and Orthopedics, Foot and Ankle Center, Peking University People's Hospital, 11th Xizhimen South Street, Beijing, China, 100044.
| |
Collapse
|
6
|
Breland AN, Ross MK, Fitzkee NC, Elder SH. In Silico Insights into the Inhibition of ADAMTS-5 by Punicalagin and Ellagic Acid for the Treatment of Osteoarthritis. Int J Mol Sci 2025; 26:4093. [PMID: 40362332 PMCID: PMC12071641 DOI: 10.3390/ijms26094093] [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: 03/24/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
ADAMTS-5 (aggrecanase-2) is a major metalloprotease involved in regulating the cartilage extracellular matrix. Due to its role in removing aggrecan in the progression of osteoarthritis (OA), ADAMTS-5 is often regarded as a potential therapeutic target for OA. Punicalagin (PCG), a polyphenolic ellagitannin found in pomegranate (Punica grunatum L.), and ellagic acid (EA), a hydrolytic metabolite of PCG, have been widely investigated as potential disease-modifying osteoarthritis drugs (DMOADs) due to their potent antioxidant and anti-inflammatory properties, but their interaction with ADAMTS-5 has yet to be determined. In this study, molecular docking simulations were used to predict enzyme-inhibitor binding interactions. The results suggest that both compounds may be able to bind within the active site via the formation of H bonds and interactions between the ligand's aromatic rings and hydrophobic residue in the enzyme with inhibition constants of 183.3 µM and 1.13 µM for PCG and EA, respectively. Biochemical activity against recombinant human ADAMTS-5 was assessed using a dimethylmethylene blue-based assay to determine residual sulfated glycosaminoglycan (sGAG) in porcine articular cartilage. Although its loss could not be attributed to ADAMTS-5, sGAG was effectively persevered by PCG and EA. The potential conversion of PCG to EA by enzyme-catalyzed hydrolysis activity was then investigated using liquid chromatography-mass spectroscopy to determine the potential for the use of PCG and EA as a prodrug-proactive metabolite pair in the development of drug delivery systems to arthritic synovial joints.
Collapse
Affiliation(s)
- Austen N. Breland
- Department of Agricultural & Biological Engineering, Mississippi State University, Starkville, MS 39762, USA;
| | - Matthew K. Ross
- Department of Comparative Biomedical Sciences, Mississippi State University, Starkville, MS 39762, USA;
| | - Nicholas C. Fitzkee
- Department of Chemistry, Mississippi State University, Starkville, MS 39762, USA;
| | - Steven H. Elder
- Department of Agricultural & Biological Engineering, Mississippi State University, Starkville, MS 39762, USA;
| |
Collapse
|
7
|
Colares MC, Thirupathi A, Beirão ME, Zaccaron RP, de Roch Casagrande L, Venturini LM, Mendes C, De-Pieri E, Gu Y, Machado-de-Ávila RA, Silveira PCL. Platelet-rich plasma and IL-1β antagonist receptor peptide attenuate the inflammatory process of muscle injury in wistar rats. Sci Rep 2025; 15:14219. [PMID: 40269232 PMCID: PMC12019253 DOI: 10.1038/s41598-025-98998-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/16/2025] [Indexed: 04/25/2025] Open
Abstract
Muscle injuries are frequent in sports, necessitating therapies that reduce inflammation and enhance repair. Platelet-rich plasma (PRP), derived from autologous blood, offers high concentrations of platelets and growth factors for tissue regeneration. Interleukin-1 beta (IL-1β), a pro-inflammatory cytokine, is crucial in inflammation, and its inhibition may speed up healing, but the high cost of blockers limits their use. This study evaluated the effects of leukocyte-rich PRP (LR-PRP), leukocyte-poor PRP (LP-PRP), and the peptide antagonist DAP1-2 on inflammation after muscle injury. Eighty-four rats were divided into seven groups: Control; muscle injury (MI); MI + LR-PRP; MI + LP-PRP; MI + DAP1-2; MI + LR-PRP + DAP1-2; MI + LP-PRP + DAP1-2. Muscle damage was induced via contusion of the right gastrocnemius, with treatments administered 24 h post-injury. On day five, groups were euthanized for analysis. RT-qPCR measured NF-kB and IL-1β expression, while ELISA assessed pro- and anti-inflammatory cytokines, including TNF-α and IL-6. All treated groups showed reduced inflammatory and oxidative stress markers by day five, with the PRP-LP + DAP1-2 group showing the most significant effects, indicating enhanced tissue repair.
Collapse
Affiliation(s)
- Mateus Cardoso Colares
- Faculty of Sports Science, Ningbo University, Ningbo, 315211, China
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil
| | - Anand Thirupathi
- Faculty of Sports Science, Ningbo University, Ningbo, 315211, China.
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil.
| | - Marcelo Emílio Beirão
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil
| | - Rubya Pereira Zaccaron
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil
| | - Laura de Roch Casagrande
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil
| | - Ligia Milanez Venturini
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil
| | - Carolini Mendes
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil
| | - Ellen De-Pieri
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil
| | - Yaodong Gu
- Faculty of Sports Science, Ningbo University, Ningbo, 315211, China
| | - Ricardo Andrez Machado-de-Ávila
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil
| | - Paulo Cesar Lock Silveira
- Faculty of Sports Science, Ningbo University, Ningbo, 315211, China.
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Universitário - Block S, Room 17, Criciúma, Santa Catarina State, 88806-000, Brazil.
| |
Collapse
|
8
|
Leibold CS, Hecker A, Schmaranzer F, Siebenrock KA. Bone wax can lead to foreign body reaction and local osteolysis after open femoroacetabular impingement (FAI) surgery. Arch Orthop Trauma Surg 2025; 145:224. [PMID: 40186677 PMCID: PMC11972202 DOI: 10.1007/s00402-025-05821-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
INTRODUCTION Bone wax is a haemostatic agent, widely used to prevent bleeding from bone surfaces. Despite its effectiveness in haemostatic control, it can lead to foreign body granuloma and osteolysis. Therefore, the aim of this study was to assess the rate and progress of osteolysis after surgical bone wax application. METHODS We included 425 patients between 01/2002 and 12/2006 that underwent offset correction for cam type femoroacetabular impingement with application of bone wax for homogeneous statistical cohort formation. Comparison was made to a similar cohort group undergoing offset correction without application of bone wax, including 479 patients between 01/2008 and 12/2012. Out of the study group, six hips in five patients presented with persisting pain and growing osteolysis on the X-rays in the area of the offset correction, and two underwent subsequent revision surgery. None of the patients in the cohort group presented with osteolysis. In both groups, patients who presented with persisting pain without radiological osteolysis had other determinable causes as labral tears, progressing osteoarthritis, trochanteric bursitis, and adhesions as suggested source of the pain. We measured the relative area of the osteolysis where present (area of osteolysis/area of femoral head in %) on lateral radiographs on the first postoperative X-rays and latest follow-up X-rays, with a mean follow-up time of 8.6 ± 2.5 years (range, 5-13 years). Histologic samples were taken at revision surgery. RESULTS The relative area of osteolysis increased in all hips from a directly postoperative median of 5.5% ± 2.7% (2.3-10.7%) to 11.2% ± 3.9% (7.1-17.3%) at last follow-up. In patients undergoing revision surgery for osteolysis, remaining wax as a foreign material with attached multinucleated giant cells and abundant mononuclear cells was detected histologically. CONCLUSION The intra-articular use of bone wax should be approached with caution and with awareness of the possible complications. TRIAL REGISTRATION NUMBER KEK 2018-00078, registered April 2018. LEVEL OF EVIDENCE level IV, retrospective case series.
Collapse
Affiliation(s)
- Christiane Sylvia Leibold
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University hospital, University of Bern, Freiburgstrasse, Bern, 3010, Switzerland.
| | - Andreas Hecker
- ArthroClinic Bern, Lindenhofspital, Bremgartenstrasse 117, 3012, Bern, Switzerland.
| | - Florian Schmaranzer
- Department of Diagnostic-, Interventional-, and Pediatric Radiology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse, Bern, 3010, Switzerland
- Faculty of Medicine, Department of Radiology, Balgrist University Hospital, University of Zürich, Forchstrasse 340, 8008, Zürich, Switzerland
| | - Klaus Arno Siebenrock
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University hospital, University of Bern, Freiburgstrasse, Bern, 3010, Switzerland.
| |
Collapse
|
9
|
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
|
10
|
Zhu Z, Tu B, Peng C, Xu X, Lu P, Ning R. Integrated bioinformatics and clinical data identify three novel biomarkers for osteoarthritis diagnosis and synovial immune. Sci Rep 2025; 15:10987. [PMID: 40164659 PMCID: PMC11958655 DOI: 10.1038/s41598-025-95837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease that can be aggravated by synovitis and synovial immune disorders (SID). However, the role of synovial SID-related genes in OA synovium remains poorly understood. OA synovial and peripheral blood datasets were obtained from the GEO database ( https://www.ncbi.nlm.nih.gov/ ). Immune-related genes ( https://reactome.org/ ) showing differential expression in peripheral blood were identified as immune disorder genes. Subsequently, differentially expressed immune disorder genes in OA synovium were further identified as SID genes. The Venn diagram, random forest, SVM-RFE algorithm, and multivariate analysis were employed to determine SID-related hub genes in OA synovium. Using the identified hub genes, we constructed and validated a diagnostic model for predicting OA occurrence. The correlation between hub gene expression and immune-related modules was explored using CIBERSORT and MCP-counter analyses. We identified three SID-related hub genes (ACAT1, SPHK1, and ACACB) in OA synovium. The diagnostic model incorporating these hub genes demonstrated reliable predictive accuracy (AUC = 0.939). Through qPCR analysis, we quantitated the expression levels of the hub genes and confirmed that three hub genes could serve as novel biomarkers for OA patients (AUC = 0.960). Furthermore, we observed a significant correlation between the expression of these hub genes and immune cell infiltration, as well as inflammatory cytokine levels in OA synovium. Our findings suggest that three SID-related hub genes have the potential to serve as diagnostic biomarkers for OA patients. These genes are associated with immune disorder and contribute to immune alterations within the OA synovium.
Collapse
Affiliation(s)
- Zheng Zhu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Bizhi Tu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Cheng Peng
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Xun Xu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Peizhi Lu
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China
| | - Rende Ning
- Department of Orthopedics, Hefei First People's Hospital, Anhui Medical University, 390 Huaihe Road, Hefei, 230061, Anhui, China.
| |
Collapse
|
11
|
Fan W, Zhao W, Hu R, Wei C, Sun L, Hou T, Li R, Sun Q, Liu C. Insights From m6A RNA Methylation: Biomarkers for Diagnosis of Acute Myocardial Infarction. J Inflamm Res 2025; 18:3589-3605. [PMID: 40093945 PMCID: PMC11910935 DOI: 10.2147/jir.s512476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose Acute myocardial infarction (AMI) is a major contributor to death. The purpose of this study is to explore circulating biomarkers for AMI diagnosis from the perspectives of immunological microenvironment and N6-methyladenosine (m6A) RNA methylation regulation. Patients and Methods The GSE59867 dataset was used to download platform and probe data for conducting differential analysis of m6A regulators. A diagnostic nomogram was created utilizing the random-forest method and evaluated for predictive power. m6A-related gene patterns were identified, and their immune microenvironment characteristics were analyzed. Peripheral blood samples were obtained for validation in patient-based investigations using RT-qPCR. The association between m6A regulators and clinical parameters was examined via Spearman correlation analysis. Results With a predictive nomogram model developed using key m6A regulators, two distinct m6A subtypes were identified, showing significant variations in infiltrating immunocyte abundance. In confirmation of the model prediction, examination of patient blood identified METTL3, WTAP, RBM15, ALKBH5, FTO, and FMR1 as novel circulating biomarkers for AMI diagnosis. METTL3 and FTO were identified as promising biomarkers for AMI given that they showed a positive correlation with left ventricular ejection fraction. Conclusion The study identified six m6A regulators as circulating biomarkers for AMI diagnosis and suggested a potential role for m6A-mediated immune cell infiltration in the pathogenesis of AMI.
Collapse
Affiliation(s)
- Wenjun Fan
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, 310053, People’s Republic of China
| | - Wenbin Zhao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, 310053, People’s Republic of China
| | - Renjie Hu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, 310053, People’s Republic of China
| | - Chen Wei
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei, 067000, People’s Republic of China
| | - Lixian Sun
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei, 067000, People’s Republic of China
| | - Tong Hou
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, 310053, People’s Republic of China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, 310053, People’s Republic of China
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, 310053, People’s Republic of China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, 310053, People’s Republic of China
| |
Collapse
|
12
|
Suarez-Ahedo C, Martinez-Armenta C, Martínez-Gómez LE, González-Mendoza O, Hernández Rocha MDJ, Martínez-Nava GA, Pineda C, López-Reyes A. Effect of Plasminogen Activator Inhibitor-1 on extracellular matrix homeostasis in scaffold-free spheroids from human chondrocytes. REUMATOLOGIA CLINICA 2025; 21:501815. [PMID: 40316399 DOI: 10.1016/j.reumae.2025.501815] [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: 10/15/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 05/04/2025]
Abstract
INTRODUCTION New trends in osteoarthritis research focus on the use of biological therapy; in this context, the use of Plasminogen Activator Inhibitor-1 (PAI-1) is considered a potential therapeutic strategy to prevent extracellular matrix (ECM) degradation in osteoarthritis (OA) management. However, in vitro studies have not demonstrated its effect on the expression of ECM homeostasis-related genes. METHODS Human OA cartilage-derived chondrocytes were used to generate scaffold-free spheroids under hypoxia conditions. The spheroids were exposed to PAI-1 for 24h, and cell viability was measured. Then qRT-PCR was used to analyze the expression of ECM components and degradative enzymes, including COL2A1, SOX9, ACAN, COL1A1, MMP3, MMP9, MMP13, ADAMTS4, ADAMTS5, TIMP1, TIMP2, TIMP3, uPA and tPA. RESULTS PAI-1 treatment consistently maintained cell viability and chondrocyte spheroid integrity. At the 50ng/mL concentration, PAI-1 increased the gene expression of COL2A1 and reduced SOX9, ACAN, MMP3, MMP9, TIMP2, and tPA. Moreover, the functional COL2A1/COL1A1 ratio was significantly increased in PAI-1-treated spheroids. CONCLUSION Our results suggest that PAI-1 treatment exerts a complex and multifaceted influence on spheroids' ECM. While it supports matrix integrity by reducing the gene expression of ECM remodeling enzymes, such as MMPs and ADAMTS5, it also induces unfavorable changes in chondrogenesis-related marker genes, such as SOX9 and ACAN. These findings indicate that the cellular response to PAI-1 is not unidirectional, warranting further investigation to understand its precise biological implications.
Collapse
Affiliation(s)
- Carlos Suarez-Ahedo
- National Rehabilitation Institute of Mexico, Geroscience Laboratory, Mexico City, Mexico; National Rehabilitation Institute of Mexico, Adult Hip and Knee Reconstruction Department, Mexico City, Mexico; American Hip Institute, Chicago, IL, USA
| | | | - Laura E Martínez-Gómez
- National Rehabilitation Institute of Mexico, Geroscience Laboratory, Mexico City, Mexico
| | | | | | | | - Carlos Pineda
- National Rehabilitation Institute of Mexico, Mexico City, Mexico
| | - Alberto López-Reyes
- National Rehabilitation Institute of Mexico, Geroscience Laboratory, Mexico City, Mexico.
| |
Collapse
|
13
|
Liu T, Jin C, Sun J, Zhu L, Wang C, Xiao F, Liu X, Lv L, Yang X, Zhou W, Tan C, Wang X, Wei W. Paroxetine alleviates dendritic cell and T lymphocyte activation via GRK2-mediated PI3K-AKT signaling in rheumatoid arthritis. Chin Med J (Engl) 2025; 138:441-451. [PMID: 38879805 PMCID: PMC11845201 DOI: 10.1097/cm9.0000000000003165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND G protein-coupled receptor kinase 2 (GRK2) could participate in the regulation of diverse cells via interacting with non-G-protein-coupled receptors. In the present work, we explored how paroxetine, a GRK2 inhibitor, modulates the differentiation and activation of immune cells in rheumatoid arthritis (RA). METHODS The blood samples of healthy individuals and RA patients were collected between July 2021 and March 2022 from the First Affiliated Hospital of Anhui Medical University. C57BL/6 mice were used to induce the collagen-induced arthritis (CIA) model. Flow cytometry analysis was used to characterize the differentiation and function of dendritic cells (DCs)/T cells. Co-immunoprecipitation was used to explore the specific molecular mechanism. RESULTS In patients with RA, high expression of GRK2 in peripheral blood lymphocytes, accompanied by the increases of phosphatidylinositol 3 kinase (PI3K), protein kinase B (AKT), and mammalian target of rapamycin (mTOR). In animal model, a decrease in regulatory T cells (T regs ), an increase in the cluster of differentiation 8 positive (CD8 + ) T cells, and maturation of DCs were observed. Paroxetine, when used in vitro and in CIA mice, restrained the maturation of DCs and the differentiation of CD8 + T cells, and induced the proportion of T regs . Paroxetine inhibited the secretion of pro-inflammatory cytokines, the expression of C-C motif chemokine receptor 7 in DCs and T cells. Simultaneously, paroxetine upregulated the expression of programmed death ligand 1, and anti-inflammatory cytokines. Additionally, paroxetine inhibited the PI3K-AKT-mTOR metabolic pathway in both DCs and T cells. This was associated with a reduction in mitochondrial membrane potential and changes in the utilization of glucose and lipids, particularly in DCs. Paroxetine reversed PI3K-AKT pathway activation induced by 740 Y-P (a PI3K agonist) through inhibiting the interaction between GRK2 and PI3K in DCs and T cells. CONCLUSION Paroxetine exerts an immunosuppressive effect by targeting GRK2, which subsequently inhibits the metabolism-related PI3K-AKT-mTOR pathway of DCs and T cells in RA.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230022, China
| | - Chao Jin
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Jing Sun
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230022, China
| | - Lina Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Chun Wang
- Institute of Clinical Pharmacology, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, China
| | - Feng Xiao
- Institute of Clinical Pharmacology, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, China
| | - Xiaochang Liu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Liying Lv
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Xiaoke Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Wenjing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230022, China
| | - Chao Tan
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230022, China
| | - Xianli Wang
- Department of Pharmacy, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, China
| |
Collapse
|
14
|
Lascelles BDX, Ponnala R, Kamerling SG, Williams T. Proteomic profiling of serum in cats with naturally occurring degenerative joint disease and co-morbid conditions. FRONTIERS IN PAIN RESEARCH 2025; 6:1501932. [PMID: 39968160 PMCID: PMC11832531 DOI: 10.3389/fpain.2025.1501932] [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: 09/25/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Degenerative joint disease (DJD) occurs very commonly in cats and can be associated with pain. Almost 70% of cats with DJD-associated pain suffer the co-morbidity of chronic kidney disease (CKD). There are currently very limited treatment or management options. A greater understanding of the systems biology of DJD, DJD-associated pain, and CKD may contribute to identifying disease specific biomarkers and relevant targets for the development of therapeutics for the control of these conditions in cats, and help inform human pain therapeutic development. Methods Using mass spectrometry-based proteomic profiling of the serum of 200 highly phenotyped cats with varying burdens of DJD, pain, and CKD, we identified significant individual proteins and pathways. Results Functional pathway analysis, based on differentially abundant proteins across individual disease states (DJD, pain, CKD), identified pathways playing a role in DJD and DJD-associated pain including acute phase response signaling, LXR/RXR and FXR/RXR activation and the complement system. With the added co-morbidity of CKD, similar pathways were identified, with the addition of IL-12 signaling and production in macrophages. Discussion We identified differentially abundant proteins associated with DJD, pain and CKD and future work should evaluate these proteins as potential biomarkers of disease (individually or as clusters). Further, these data could be leveraged to identify novel therapeutic targets to address the gap in our ability to manage DJD, pain, and CKD in cats. Given that our work was in cats with naturally occurring DJD, these results may have translational applicability to human health.
Collapse
Affiliation(s)
- B. Duncan X. Lascelles
- Translational Research in Pain, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, NC, United States
- Thurston Arthritis Center, UNC School of Medicine, Chapel Hill, NC, United States
- Center for Translational Pain Research, Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Rakesh Ponnala
- Veterinary Medicine Research & Development, Zoetis, Kalamazoo, MI, United States
| | - Steven G. Kamerling
- Veterinary Pharmacology Consultant, Veterinary Medicine Research & Development, Zoetis, Kalamazoo, MI, United States
| | - Tracey Williams
- Veterinary Medicine Research & Development, Zoetis, Kalamazoo, MI, United States
| |
Collapse
|
15
|
Hsieh RZ, Huang KC, Su YP, Shi CS, Chang SF. The Potential Role of Bone Morphogenetic Protein-2/-4 in Excessive Mechanical Overloading-Initiated Joint Degeneration. J Cell Physiol 2025; 240:e31509. [PMID: 39710977 DOI: 10.1002/jcp.31509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024]
Abstract
Excessive mechanical overloading of articular cartilage caused by excessive exercise or severe trauma is considered a critical trigger in the development of osteoarthritis (OA). However, the available clinical theranostic molecular targets and underlying mechanisms still require more elucidation. Here, we aimed to examine the possibility that bone morphogenetic proteins (BMPs) serve as molecular targets in rat cartilages and human chondrocytes under conditions of excessive mechanical overloading. Two rat models involving high-intensity running training and surgery for destabilization of medial meniscus, along with a cell model subjected to cyclic tensile strain, were established to simulate and investigate excessive mechanical overloading effects on cartilages/chondrocytes. We employed various methods, including immunohistochemistry, real-time polymerase chain reaction, western blot analysis, and enzyme-linked immunosorbent assay, to evaluate the expression, secretion, phosphorylation, and nuclear translocation of mRNA/proteins in cartilages and chondrocytes. Our findings revealed a simultaneous upregulation of BMP-2 and downregulation of BMP-4 in degenerated and inflamed cartilages and chondrocytes under excessive mechanical overloading. Furthermore, toll-like receptor 2 and nuclear factor kappa B-p50/p65 subunits signaling were identified as regulators governing this distinct expression pattern. Treatment with recombinant BMP-2 and/or BMP-4 proteins significantly ameliorated cartilage degeneration and chondrocyte inflammation induced by excessive mechanical overloading. These results strongly suggest that BMP-2 upregulation and BMP-4 downregulation might represent mechanisms for self-rescue and degeneration in damaged cartilage/chondrocytes, respectively. Our findings advance new insights that BMP-2/-4 might be potential molecular targets for excessive mechanical overloading-caused OA development and should be taken into account in future clinical applications.
Collapse
Affiliation(s)
- Rong-Ze Hsieh
- Department of Medical Research and Development, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuo-Chin Huang
- Department of Orthopaedics, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Ping Su
- Department of Orthopaedics and Traumatology, Veterans General Hospital, Taipei, Taiwan
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Sheng Shi
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Shun-Fu Chang
- Department of Medical Research and Development, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Center for General Education, Chiayi Chang Gung University of Science and Technology, Chiayi, Taiwan
| |
Collapse
|
16
|
Walton BL, Shattuck-Brandt R, Hamann CA, Tung VW, Colazo JM, Brand DD, Hasty KA, Duvall CL, Brunger JM. A programmable arthritis-specific receptor for guided articular cartilage regenerative medicine. Osteoarthritis Cartilage 2025; 33:231-240. [PMID: 39706287 PMCID: PMC12019866 DOI: 10.1016/j.joca.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE Investigational cell therapies have been developed as disease-modifying agents for the treatment of osteoarthritis (OA), including those that inducibly respond to inflammatory factors driving OA progression. However, dysregulated inflammatory cascades do not specifically signify the presence of OA. Here, we deploy a synthetic receptor platform that regulates cell behaviors in an arthritis-specific fashion to confine transgene expression to sites of cartilage degeneration. DESIGN A single-chain variable fragment specific for type II collagen (CII) that is exposed in damaged cartilage was used to produce a synthetic Notch (synNotch) receptor that enables "CII-synNotch" mesenchymal stromal cells (MSCs) to recognize degraded cartilage. Artificial signaling induced by both CII-treated culture surfaces and primary tissues was measured via fluorescence and luminescence assays. Separate studies measured the ability of CII-synNotch to govern cartilage anabolic activity of MSCs. Finally, a co-culture with ATDC5 chondrocytes was used to determine whether CII-synNotch MSCs can protect chondrocytes against deleterious effects of pro-inflammatory interleukin-1 in a CII-dependent manner. RESULTS CII-synNotch MSCs are highly and selectively responsive to CII, but not type I collagen, as measured by luminescence assays, fluorescence microscopy, and concentrations of secreted transgene products in culture media. CII-synNotch cells exhibit the capacity to distinguish between healthy and damaged cartilage tissue and constrain transgene expression to regions of exposed CII fibers. Receptor-regulated production of cartilage anabolic and anti-inflammatory transgenes was effective to mediate cartilage regenerative functions. CONCLUSION This work demonstrates proof-of-concept that the synNotch platform guides MSCs for spatially regulated, disease-dependent delivery of OA-relevant biologic drugs.
Collapse
Affiliation(s)
- Bonnie L Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | | | - Catherine A Hamann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Victoria W Tung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - David D Brand
- Research Service, Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN 38105, USA
| | - Karen A Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis VA Medical Center, Memphis, TN 38105, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA; Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA; Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37212, USA.
| |
Collapse
|
17
|
Xi Y, Wang Z, Wei Y, Xiao N, Duan L, Zhao T, Zhang X, Zhang L, Wang J, Li Z, Qin D. Gut Microbiota and Osteoarthritis: From Pathogenesis to Novel Therapeutic Opportunities. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:43-66. [PMID: 39880660 DOI: 10.1142/s0192415x2550003x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Osteoarthritis (OA) is the most common chronic degenerative joint disease, characterized by cartilage damage, synovial inflammation, subchondral bone sclerosis, marginal bone loss, and osteophyte development. Clinical manifestations include inflammatory joint pain, swelling, osteophytes, and limitation of motion. The pathogenesis of osteoarthritis has not yet been fully uncovered. With ongoing research, however, it has been gradually determined that OA is not caused solely by mechanical injury or aging, but rather involves chronic low-grade inflammation, metabolic imbalances, dysfunctional adaptive immunity, and alterations in central pain processing centers. The main risk factors for OA include obesity, age, gender, genetics, and sports injuries. In recent years, extensive research on gut microbiota has revealed that gut dysbiosis is associated with some common risk factors for OA, and that it may intervene in its pathogenesis through both direct and indirect mechanisms. Therefore, gut flora imbalance as a pathogenic factor in OA has become a hotspot topic of research, with potential therapeutic connotations. In this paper, we review the role of the gut microbiota in the pathogenesis of OA, describe its relationship with common OA risk factors, and address candidate gut microbiota markers for OA diagnosis. In addition, with focus on OA therapies, we discuss the effects of direct and indirect interventions targeting the gut microbiota, as well as the impact of gut bacteria on the efficacy of OA drugs.
Collapse
Affiliation(s)
- Yujiang Xi
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
- United Graduate School, China Academy of Chinese Medical Sciences, Suzhou Jiangsu 215000, P. R. China
| | - Zhifeng Wang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
| | - Yuanyuan Wei
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
| | - Niqin Xiao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
| | - Li Duan
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
| | - Ting Zhao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
| | - Xiaoyu Zhang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
| | - Liping Zhang
- Southern Central Hospital of Yunnan Province, Mengzi Honghe 661100, P. R. China
| | - Jian Wang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
| | - Zhaofu Li
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
| | - Dongdong Qin
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming Yunnan 650500, P. R. China
| |
Collapse
|
18
|
Figueroa-Valdés AI, Luz-Crawford P, Herrera-Luna Y, Georges-Calderón N, García C, Tobar HE, Araya MJ, Matas J, Donoso-Meneses D, de la Fuente C, Cuenca J, Parra E, Lillo F, Varela C, Cádiz MI, Vernal R, Ortloff A, Nardocci G, Castañeda V, Adasme-Vidal C, Kunze-Küllmer M, Hidalgo Y, Espinoza F, Khoury M, Alcayaga-Miranda F. Clinical-grade extracellular vesicles derived from umbilical cord mesenchymal stromal cells: preclinical development and first-in-human intra-articular validation as therapeutics for knee osteoarthritis. J Nanobiotechnology 2025; 23:13. [PMID: 39806427 PMCID: PMC11730155 DOI: 10.1186/s12951-024-03088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Osteoarthritis (OA) is a joint disease characterized by articular cartilage degradation. Persistent low-grade inflammation defines OA pathogenesis, with crucial involvement of pro-inflammatory M1-like macrophages. While mesenchymal stromal cells (MSC) and their small extracellular vesicles (sEV) hold promise for OA treatment, achieving consistent clinical-grade sEV products remains a significant challenge. This study aims to develop fully characterized, reproducible, clinical-grade batches of sEV derived from umbilical cord (UC)-MSC for the treatment of OA while assessing its efficacy and safety. Initially, a standardized, research-grade manufacturing protocol was established to ensure consistent sEV production. UC-MSC-sEV characterization under non-cGMP conditions showed consistent miRNA and protein profiles, suggesting their potential for standardized manufacturing. In vitro studies evaluated the efficacy, safety, and potency of sEV; animal studies confirmed their effectiveness and safety. In vitro, UC-MSC-sEV polarized macrophages to an anti-inflammatory M2b-like phenotype, through STAT1 modulation, indicating their potential to create an anti-inflammatory environment in the affected joints. In silico studies confirmed sEV's immunosuppressive signature through miRNA and proteome analysis. In an OA mouse model, sEV injected intra-articularly (IA) induced hyaline cartilage regeneration, validated by histological and μCT analyses. The unique detection of sEV signals within the knee joint over time highlights its safety profile by confirming the retention of sEV in the joint. The product development of UC-MSC-sEV involved refining, standardizing, and validating processes in compliance with GMP standards. The initial assessment of the safety of the clinical-grade product via IA administration in a first-in-human study showed no adverse effects after a 12 month follow-up period. These results support the progress of this sEV-based therapy in an early-phase clinical trial, the details of which are presented and discussed in this work. This study provides data on using UC-MSC-sEV as local therapy for OA, highlighting their regenerative and anti-inflammatory properties and safety in preclinical and a proof-of-principle clinical application.
Collapse
Affiliation(s)
- Aliosha I Figueroa-Valdés
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Nicolás Georges-Calderón
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Hugo E Tobar
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - José Matas
- Centro de Terapia Celular, Clínica Universidad de los Andes, Santiago, Chile
- Departmento de Cirugía Ortopédica, Clínica Universidad de los Andes, Santiago, Chile
| | - Darío Donoso-Meneses
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | | | - Jimena Cuenca
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Eliseo Parra
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Fernando Lillo
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Cristóbal Varela
- Departmento de Radiología, Clínica Universidad de los Andes, Santiago, Chile
| | - María Ignacia Cádiz
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Rolando Vernal
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Gino Nardocci
- Laboratorio de Biología Molecular y Bioinformática, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Verónica Castañeda
- Laboratorio de Biología Molecular y Bioinformática, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Catalina Adasme-Vidal
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Maximiliano Kunze-Küllmer
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- Cells for Cells, Santiago, Chile
- EVast Bio, Miami, FL, USA
| | - Yessia Hidalgo
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Francisco Espinoza
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Centro de Terapia Celular, Clínica Universidad de los Andes, Santiago, Chile
- Departmento de Reumatología, Clínica Universidad de los Andes, Santiago, Chile
| | - Maroun Khoury
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile.
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cells for Cells, Santiago, Chile.
- EVast Bio, Miami, FL, USA.
| | - Francisca Alcayaga-Miranda
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile.
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cells for Cells, Santiago, Chile.
| |
Collapse
|
19
|
Meléndez-Oliva E, Martínez-Pozas O, Sinatti P, Martín Carreras-Presas C, Cuenca-Zaldívar JN, Turroni S, Sánchez Romero EA. Relationship Between the Gut Microbiome, Tryptophan-Derived Metabolites, and Osteoarthritis-Related Pain: A Systematic Review with Meta-Analysis. Nutrients 2025; 17:264. [PMID: 39861394 PMCID: PMC11767305 DOI: 10.3390/nu17020264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is the most prevalent form of arthritis and affects over 528 million people worldwide. Degenerative joint disease involves cartilage degradation, subchondral bone remodeling, and synovial inflammation, leading to chronic pain, stiffness, and impaired joint function. Initially regarded as a "wear and tear" condition associated with aging and mechanical stress, OA is now recognized as a multifaceted disease influenced by systemic factors such as metabolic syndrome, obesity, and chronic low-grade inflammation. Recent studies have focused on the gut-joint axis to investigate how the gut microbiome modulates inflammation and pain in OA. MATERIALS AND METHODS A systematic review was conducted following the PRISMA guidelines and was registered with PROSPERO (CRD42024556265). This review included studies involving adults with symptomatic OA and analyzed the relationship between the gut microbiome and OA-related pain. Randomized and non-randomized clinical trials, case reports, editorials, and pilot studies were excluded. Searches were performed in PubMed, Cochrane Library, and Web of Science without publication date restrictions, and filtered for "observational studies". The study selection and data extraction were performed by two independent researchers, and the risk of bias was assessed using appropriate tools. RESULTS Five observational studies were included in the systematic review, and three were included in the meta-analysis. Two studies reported an association between different tryptophan metabolites and pain levels in patients with OA. Two other studies demonstrated a correlation between lipopolysaccharide levels and pain in OA. A fifth study confirmed the relationship between Streptococcus relative abundance of Streptococcus spp. and knee pain. These results were not supported by a meta-analysis, which found no significant association between the presence of pain in OA and the presence of bacilli of the genus Streptococcus or plasma markers of the tryptophan pathway. CONCLUSIONS Current evidence indicates a potential link between gut microbiome dysbiosis and OA-related pain. However, methodological limitations preclude definitive conclusions. Further research using advanced techniques and larger cohorts is needed to validate and extend these findings and elucidate the underlying mechanisms. Targeted manipulation of the gut microbiome may be a valuable strategy for pain management in OA patients.
Collapse
Affiliation(s)
- Erika Meléndez-Oliva
- Grupo de Investigación en Dietética Aplicada, Nutrición y Composición Corporal (DANuC), Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain;
- Grupo de Investigación en Calidad de Vida y Salud, Departamento de Ciencias de la Salud, Universidad Europea de Valencia, 03016 Alicante, Spain
- Physiotherapy and Orofacial Pain Working Group, Sociedad Española de Disfunción Craneomandibular y Dolor Orofacial (SEDCYDO), 28009 Madrid, Spain
- Interdisciplinary Research Group on Musculoskeletal Disorders, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain;
| | - Oliver Martínez-Pozas
- Physiotherapy and Orofacial Pain Working Group, Sociedad Española de Disfunción Craneomandibular y Dolor Orofacial (SEDCYDO), 28009 Madrid, Spain
- Interdisciplinary Research Group on Musculoskeletal Disorders, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain;
- Escuela Internacional de Doctorado, Faculty of Health Sciences, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain
| | - Pierluigi Sinatti
- Interdisciplinary Research Group on Musculoskeletal Disorders, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain;
- IPPOCRATE Centro Medico Specialistico, Via La Spezia 38, 00055 Ladispoli, Italy
| | - Carmen Martín Carreras-Presas
- Special Care Dentistry, Oral Medicine and Quality of Life Research Gorup (SOUL), Oral Medicine Unit, Faculty of Dentistry, European University of Madrid, 28670 Madrid, Spain;
| | - Juan Nicolás Cuenca-Zaldívar
- Interdisciplinary Research Group on Musculoskeletal Disorders, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain;
- Grupo de Investigación en Fisioterapia y Dolor, Departamento de Fisioterapia, Facultad de Enfermería y Fisioterapia, Universidad de Alcalá, 28801 Alcalá de Henares, Spain
- Research Group in Nursing and Health Care, Puerta de Hierro Health Research Institute-Segovia de Arana (IDIPHISA), 28222 Majadahonda, Spain
- Physical Therapy Unit, Primary Health Care Center «El Abajón», 28231 Las Rozas de Madrid, Spain
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| | - Eleuterio A. Sánchez Romero
- Grupo de Investigación en Calidad de Vida y Salud, Departamento de Ciencias de la Salud, Universidad Europea de Valencia, 03016 Alicante, Spain
- Physiotherapy and Orofacial Pain Working Group, Sociedad Española de Disfunción Craneomandibular y Dolor Orofacial (SEDCYDO), 28009 Madrid, Spain
- Interdisciplinary Research Group on Musculoskeletal Disorders, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain;
- Research Group in Nursing and Health Care, Puerta de Hierro Health Research Institute-Segovia de Arana (IDIPHISA), 28222 Majadahonda, Spain
- Faculty of Medicine, Health and Sports, Department of Physiotherapy, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain
| |
Collapse
|
20
|
Wu Z, Xu K, Chen M, Wang S, Ma Y. Protective role of Yougui Yin in experimental knee osteoarthritis: From the perspective of macrophage polarization. Prostaglandins Other Lipid Mediat 2025; 176:106940. [PMID: 39674342 DOI: 10.1016/j.prostaglandins.2024.106940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Knee osteoarthritis (KOA) refers to a prevalent musculoskeletal disorder, frequently complicated by substantial pain and physical disability. Yougui Yin (YGY) is a classic Chinese herbal mixture which has demonstrated potential in treating KOA. Considering that, its cryptic mechanism warrants to be deciphered, which is the subject of our present research. In vivo, H&E staining, Alcian blue staining and Masson staining assessed the histomorphology. Commercial kits and ELISA evaluated oxidative stress markers. ELISA also assayed serum inflammatory cytokines. TUNEL staining appraised apoptosis. Western blotting examined cartilage matrix degradation, apoptotic and NLRP3 inflammasome proteins. Immunofluorescence assay estimated macrophage polarization. In vitro, ELISA assayed oxidative stress markers and inflammatory cytokines. Immunofluorescence and flow cytometry assay estimated macrophage polarization. MTT and flow cytometry assays severally measured cell viability and apoptosis. DCFH-DA probe detected ROS formation. RT-qPCR and Western blotting examined chondrocyte markers, apoptotic and pyroptotic genes. YGY significantly eased the histomorphological damage, apoptosis and pyroptosis in the cartilage tissues of KOA mice. Besides, YGY exerted anti-oxidant and anti-inflammatory activities and drove M1-to-M2 polarization of macrophages both in vitro and in vivo. Further, the co-culture of macrophages treated by LPS and serum containing YGY improved the viability, eliminated the apoptosis, pyroptosis, inflammation, oxidative stress and cartilage degradation in TNF-α-exposed chondrocytes co-cultured with LPS-intervened macrophages. Overall, YGY might mediate macrophage polarization to impede the advancement of KOA.
Collapse
Affiliation(s)
- Zhongqing Wu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Department of Orthopaedics, the First People's Hospital of Huzhou, Huzhou 313000, China; Department of Orthopaedics, Huzhou Wuxing District People's Hospital and Huzhou Wuxing District Maternal and Child Health Hospital, Huzhou 313008, China
| | - Kanna Xu
- Emergency Department, the First People's Hospital of Huzhou, Huzhou 313000, China
| | - Minchang Chen
- Department of Orthopaedics, Huzhou Wuxing District People's Hospital and Huzhou Wuxing District Maternal and Child Health Hospital, Huzhou 313008, China
| | - Shihao Wang
- Department of Orthopaedics, Huzhou Wuxing District People's Hospital and Huzhou Wuxing District Maternal and Child Health Hospital, Huzhou 313008, China
| | - Yong Ma
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
21
|
van Kooten NJT, Blom AB, Teunissen van Manen IJ, Theeuwes WF, Roth J, Gorris MAJ, Walgreen B, Sloetjes AW, Helsen MM, Vitters EL, van Lent PLEM, Koëter S, van der Kraan PM, Vogl T, van den Bosch MHJ. S100A8/A9 drives monocytes towards M2-like macrophage differentiation and associates with M2-like macrophages in osteoarthritic synovium. Rheumatology (Oxford) 2025; 64:332-343. [PMID: 38216750 PMCID: PMC11701306 DOI: 10.1093/rheumatology/keae020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/04/2023] [Accepted: 12/21/2023] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVES Macrophages are key orchestrators of the osteoarthritis (OA)-associated inflammatory response. Macrophage phenotype is dependent on environmental cues like the inflammatory factor S100A8/A9. Here, we investigated how S100A9 exposure during monocyte-to-macrophage differentiation affects macrophage phenotype and function. METHODS OA synovium cellular composition was determined using flow cytometry and multiplex immunohistochemistry. Healthy donor monocytes were differentiated towards M1- and M2-like macrophages in the presence of S100A9. Macrophage markers were measured using flow cytometry, and phagocytic activity was determined using pHrodo Red Zymosan A BioParticles. Gene expression was determined using qPCR. Protein secretion was measured using Luminex multianalyte analysis and ELISA. RESULTS Macrophages were the dominant leucocyte subpopulation in OA synovium. They mainly presented with an M2-like phenotype, although the majority also expressed M1-like macrophage markers. Long-term exposure to S100A9 during monocyte-to-macrophage differentiation increased M2-like macrophage markers CD163 and CD206 in M1-like and M2-like differentiated cells. In addition, M1-like macrophage markers were increased in M1-like, but decreased in M2-like differentiated macrophages. In agreement with this mixed phenotype, S100A9 stimulation modestly increased expression and secretion of pro-inflammatory markers and catabolic enzymes, but also increased expression and secretion of anti-inflammatory/anabolic markers. In accordance with the upregulation of M2-like macrophage markers, S100A9 increased phagocytic activity. Finally, we indeed observed a strong association between S100A8 and S100A9 expression and the M2-like/M1-like macrophage ratio in end-stage OA synovium. CONCLUSION Chronic S100A8/A9 exposure during monocyte-to-macrophage differentiation favours differentiation towards an M2-like macrophage phenotype. The properties of these cells could help explain the catabolic/anabolic dualism in established OA joints with low-grade inflammation.
Collapse
Affiliation(s)
- Nienke J T van Kooten
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
- Orthopedics, Canisius Wilhelmina Ziekenhuis, Nijmegen, The Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Wessel F Theeuwes
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Mark A J Gorris
- Medical BioSciences, Radboud university medical center, Nijmegen, The Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud university medical center, Nijmegen, The Netherlands
| | - Birgitte Walgreen
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Annet W Sloetjes
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Monique M Helsen
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Elly L Vitters
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Sander Koëter
- Orthopedics, Canisius Wilhelmina Ziekenhuis, Nijmegen, The Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | | |
Collapse
|
22
|
Andrei C, Mihai DP, Nitulescu GM, Nitulescu G, Zanfirescu A. Modulating Autophagy in Osteoarthritis: Exploring Emerging Therapeutic Drug Targets. Int J Mol Sci 2024; 25:13695. [PMID: 39769455 PMCID: PMC11727697 DOI: 10.3390/ijms252413695] [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: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by the breakdown of cartilage and the subsequent inflammation of joint tissues, leading to pain and reduced mobility. Despite advancements in symptomatic treatments, disease-modifying therapies for OA remain limited. This narrative review examines the dual role of autophagy in OA, emphasizing its protective functions during the early stages and its potential to contribute to cartilage degeneration in later stages. By delving into the molecular pathways that regulate autophagy, this review highlights its intricate interplay with oxidative stress and inflammation, key drivers of OA progression. Emerging therapeutic strategies aimed at modulating autophagy are explored, including pharmacological agents such as AMP kinase activators, and microRNA-based therapies. Preclinical studies reveal encouraging results, demonstrating that enhancing autophagy can reduce inflammation and decelerate cartilage degradation. However, the therapeutic benefits of autophagy modulation depend on precise, stage-specific approaches. Excessive or dysregulated autophagy in advanced OA may lead to chondrocyte apoptosis, exacerbating joint damage. This review underscores the promise of autophagy-based interventions in bridging the gap between experimental research and clinical application. By advancing our understanding of autophagy's role in OA, these findings pave the way for innovative and effective therapies. Nonetheless, further research is essential to optimize these strategies, address potential off-target effects, and develop safe, targeted treatments that improve outcomes for OA patients.
Collapse
Affiliation(s)
| | - Dragos Paul Mihai
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.A.); (G.M.N.); (G.N.); (A.Z.)
| | | | | | | |
Collapse
|
23
|
Du J, Zhou T, Zhang W, Peng W. Developing the new diagnostic model by integrating bioinformatics and machine learning for osteoarthritis. J Orthop Surg Res 2024; 19:832. [PMID: 39695788 DOI: 10.1186/s13018-024-05340-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common cause of disability among the elderly, profoundly affecting quality of life. This study aims to leverage bioinformatics and machine learning to develop an artificial neural network (ANN) model for diagnosing OA, providing new avenues for early diagnosis and treatment. METHODS From the Gene Expression Omnibus (GEO) database, we first obtained OA synovial tissue microarray datasets. Differentially expressed genes (DEGs) associated with OA were identified through utilization of the Limma package and weighted gene co-expression network analysis (WGCNA). Subsequently, protein-protein interaction (PPI) network analysis and machine learning were employed to identify the most relevant potential feature genes of OA, and ANN diagnostic model and receiver operating characteristic (ROC) curve were constructed to evaluate the diagnostic performance of the model. In addition, the expression levels of the feature genes were verified using real-time quantitative polymerase chain reaction (qRT-PCR). Finally, immune cell infiltration analysis was performed using CIBERSORT algorithm to explore the correlation between feature genes and immune cells. RESULTS The Limma package and WGCNA identified a total of 72 DEGs related to OA, of which 12 were up-regulated and 60 were down-regulated. Then, the PPI network analysis identified 21 hub genes, and three machine learning algorithms finally screened four feature genes (BTG2, CALML4, DUSP5, and GADD45B). The ANN diagnostic model was constructed based on these four feature genes. The AUC of the training set was 0.942, and the AUC of the validation set was 0.850. In addition, the qRT-PCR validation results demonstrated a significant downregulation of BTG2, DUSP5, and GADD45 mRNA expression levels in OA samples compared to normal samples, while CALML4 mRNA expression level exhibited an upregulation. Immune cell infiltration analysis revealed B cells memory, T cells gamma delta, B cells naive, Plasma cells, T cells CD4 memory resting, and NK cells The abnormal infiltration of activated cells may be related to the progression of OA. CONCLUSIONS BTG2, CALML4, DUSP5, and GADD45B were identified as potential feature genes for OA, and an ANN diagnostic model with good diagnostic performance was developed, providing a new perspective for the early diagnosis and personalized treatment of OA.
Collapse
Affiliation(s)
- Jian Du
- Department of Orthopedics, The Fourth Medical Centre, Chinese PLA General Hospital, No.51 Fucheng Road, Haidian District, Beijing, 100048, People's Republic of China
- Graduate School of Hebei North University, Zhangjiakou, 075000, China
| | - Tian Zhou
- Graduate School of Hebei North University, Zhangjiakou, 075000, China
| | - Wei Zhang
- Graduate School of Hebei North University, Zhangjiakou, 075000, China
| | - Wei Peng
- Department of Orthopedics, The Fourth Medical Centre, Chinese PLA General Hospital, No.51 Fucheng Road, Haidian District, Beijing, 100048, People's Republic of China.
| |
Collapse
|
24
|
Guo TZ, Shi X, Li X, Li WW, Wei T, Sahbaie P, McAllister TN, Angst MS, Clark JD, Kingery WS. Autoantibodies cause nociceptive sensitization in a mouse model of degenerative osteoarthritis. Pain 2024:00006396-990000000-00794. [PMID: 39835597 DOI: 10.1097/j.pain.0000000000003500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/24/2024] [Indexed: 01/22/2025]
Abstract
ABSTRACT Previous preclinical and translational studies suggest that tissue trauma related to bony fracture and intervertebral disk disruption initiates the formation of pronociceptive antibodies that support chronic musculoskeletal pain conditions. This study tested this hypothesis in the monosodium iodoacetate (MIA) mouse model of osteoarthritis (OA) and extended the findings using OA patient samples. Monosodium iodoacetate was injected unilaterally into the knees of male and female wild-type (WT) and muMT mice (lacking B cells) to induce articular cartilage damage. Repeated nociceptive behavioral testing was performed, and serum was collected for antibody isolation and passive transfer experiments. Serum antibodies collected from patients with OA were tested in MIA-treated muMT mice. Biochemical analyses were performed on knee joint tissues. Monosodium iodoacetate-treated WT mice developed chronic ipsilateral hindlimb allodynia, hyperalgesia, and unweighting, but these pain behaviors were absent in MIA-treated muMT mice, indicating that cartilage injury-induced pain is B-cell dependent. IgM accumulation was observed in the knee tissues of MIA-treated mice, and intra-articular injection of IgM from MIA-treated mice into MIA-treated muMT mice caused nociceptive sensitization. Similarly, intra-articular injection of IgM from patients with OA was pronociceptive in muMT MIA mice and control subject IgM had no effect. Monosodium iodoacetate-injected joints demonstrate elevated levels of complement component 5a (C5a) and C5a receptor blockade using intra-articular PMX-53-reduced sensitization. These data suggest that MIA-treated mice and patients with OA generate pronociceptive antibodies, and further support the pronociceptive autoimmunity hypothesis for the transition from tissue injury to chronic musculoskeletal pain.
Collapse
Affiliation(s)
- Tian-Zhi Guo
- Palo Alto Veterans Institute for Research, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Xiaoyou Shi
- Palo Alto Veterans Institute for Research, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
- Department of Anesthesiology Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Xuanying Li
- Department of Anesthesiology Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Wen-Wu Li
- Palo Alto Veterans Institute for Research, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
- Department of Anesthesiology Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Tzuping Wei
- Palo Alto Veterans Institute for Research, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Peyman Sahbaie
- Palo Alto Veterans Institute for Research, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
- Department of Anesthesiology Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Tiffany N McAllister
- Department of Anesthesiology Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Martin S Angst
- Department of Anesthesiology Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - J David Clark
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
- Department of Anesthesiology Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Wade S Kingery
- Palo Alto Veterans Institute for Research, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| |
Collapse
|
25
|
Liu B, Liu T, Li Y, Tan C. Innovative Biotherapies and Nanotechnology in Osteoarthritis: Advancements in Inflammation Control and Cartilage Regeneration. Int J Mol Sci 2024; 25:13384. [PMID: 39769149 PMCID: PMC11677281 DOI: 10.3390/ijms252413384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Osteoarthritis (OA) is among the most prevalent degenerative joint disorders worldwide, particularly affecting the aging population and imposing significant disability and economic burdens. The disease is characterized by progressive degradation of articular cartilage and chronic inflammation, with no effective long-term treatments currently available to address the underlying causes of its progression. Conventional therapies primarily manage symptoms such as pain and inflammation but fail to repair damaged tissues. Emerging biotherapies and regenerative medicine approaches offer promising alternatives by addressing cartilage repair and inflammation control at the molecular level. This review explores the recent advancements in biotherapeutic strategies, including mesenchymal stem cell (MSC) therapy, growth factors, and tissue engineering, which hold the potential for promoting cartilage regeneration and modulating the inflammatory microenvironment. Additionally, the integration of nanotechnology has opened new avenues for targeted drug delivery systems and the development of innovative nanomaterials that can further enhance the efficacy of biotherapies by precisely targeting inflammation and cartilage damage. This article concludes by discussing the current clinical applications, the ongoing clinical trials, and the future research directions necessary to confirm the safety and efficacy of these advanced therapies for OA management. With these advancements, biotherapies combined with nanotechnology may revolutionize the future of OA treatment by offering precise and effective solutions for long-term disease management and improved patient outcomes.
Collapse
Affiliation(s)
| | | | - Yanhong Li
- Department of Rheumatology and Immunology, West China Hospital of Sichuan University, Chengdu 610041, China; (B.L.); (T.L.)
| | - Chunyu Tan
- Department of Rheumatology and Immunology, West China Hospital of Sichuan University, Chengdu 610041, China; (B.L.); (T.L.)
| |
Collapse
|
26
|
Ruan S, Tang D, Luo Y, Song H. Identification of mitophagy-related biomarkers in osteoarthritis. Animal Model Exp Med 2024; 7:781-792. [PMID: 38720455 PMCID: PMC11680475 DOI: 10.1002/ame2.12416] [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: 01/16/2024] [Accepted: 03/27/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common joint disease, and existing drugs cannot cure OA, so there is an urgent need to identify new targets. Mitophagy plays an important role in OA; however, the role of mitophagy in the OA immune system is not yet clear. METHODS In this study, differential analysis and enrichment analysis were used to identify mitophagy-related genes (MRGs) with differential expression in OA and the functional pathways involved in OA. Subsequently, two machine learning methods, RF and LASSO, were used to screen MRGs with diagnostic value and construct nomograms. At the same time, the relationship between mitophagy and OA immune response was explored by immunoinfiltration analysis. RESULTS Forty-three differentially MRGs were identified in OA, of which six MRGs (GABARAPL2, PARL, GABARAPL1, JUN, RRAS, and SNX7) were associated with the diagnosis of OA. The ROC analysis results show that these 6 MRGs have high predictive accuracy in the diagnosis of OA. In immune infiltration analysis, we found that the abundance of significantly different immune cells in OA was mostly upregulated. In addition, the expression of diagnostic-related MRGs is correlated with changes in the abundance of immune cells in OA. CONCLUSION This study demonstrates that six MRGs can be used as diagnostic biomarkers. The expression of diagnostic-related MRGs is correlated with changes in the abundance of immune cells in OA. At the same time, mitophagy may affect the immune microenvironment of OA by regulating immune cells, ultimately leading to the progression of OA.
Collapse
Affiliation(s)
- Shiqiang Ruan
- Department of Orthopaedics Surgerythe First People's Hospital of Zunyi City (The Third Affiliated Hospital of Zunyi Medical University)ZunyiChina
| | - Dongxu Tang
- Department of Orthopaedics Surgerythe First People's Hospital of Zunyi City (The Third Affiliated Hospital of Zunyi Medical University)ZunyiChina
| | - Yanfei Luo
- Department of Orthopaedics Surgerythe First People's Hospital of Zunyi City (The Third Affiliated Hospital of Zunyi Medical University)ZunyiChina
| | - Hao Song
- Department of Orthopaedics Surgerythe First People's Hospital of Zunyi City (The Third Affiliated Hospital of Zunyi Medical University)ZunyiChina
| |
Collapse
|
27
|
Moon J, Cho KH, Jhun J, Choi J, Na HS, Lee JS, Lee SY, Min JK, Shetty A, Park SH, Kim SJ, Cho ML. Small heterodimer partner-interacting leucine zipper protein suppresses pain and cartilage destruction in an osteoarthritis model by modulating the AMPK/STAT3 signaling pathway. Arthritis Res Ther 2024; 26:199. [PMID: 39533324 PMCID: PMC11555939 DOI: 10.1186/s13075-024-03417-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a degenerative joint disease caused by the breakdown of joint cartilage and adjacent bone. Joint injury, being overweight, differences in leg length, high levels of joint stress, abnormal joint or limb development, and inherited factors have been implicated in the etiology of OA. In addition to physical damage to the joint, a role for inflammatory processes has been identified as well. Small heterodimer partner-interacting leucine zipper protein (SMILE) regulates transcription and many cellular functions. Among the proteins activated by SMILE is the peroxisome proliferator-activated receptor (PPAR) γ, which mediates the activities of CD4 + T helper cells, including Th1, Th2, and Th17, as well as Treg cells. PPAR-γ binds to STAT3 to inhibit its transcription, thereby suppressing the expression of the NF-κB pathway, and in turn, the expression of the inflammatory cytokines interferon (IFN), interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α, which are sub-signals of STAT3 and NF-κB. METHODS OA was induced in control C57BL/6 mice and in C57BL/6-derived SMILE-overexpressing transgenic (SMILE Tg) mice. The protein expression levels in the joint and spleen tissues were analyzed by immunohistochemistry and immunofluorescence images. In addition, flow cytometry was performed for detecting changes of the changes of immune cells. RESULTS Less cartilage damage and significantly reduced levels of OA biomarkers (MMP13, TIMP3 and MCP-1) were observed in SMILE Tg mice. Immunohistochemistry performed to identify the signaling pathway involved in the link between SMILE expression and OA revealed decreased levels of IL-1β, IL-6, TNF-α, and phosphorylated AMPK in synovial tissues as well as a significant decrease in phosphorylated STAT3 in both cartilage and synovium. Changes in systemic immune cells were investigated via flow cytometry to analyze splenocytes isolated from control and SMILE Tg mice. SMILE Tg mice had elevated proportions of CD4 + IL-4 + cells (Th2) and CD4 + CD25 + Foxp3 + cells (Treg) and a notable decrease in CD4 + IL-17 + cells (Th17). CONCLUSION Our results show that overexpressed SMILE attenuates the symptoms of OA, by increasing AMPK signaling and decreasing STAT3, thus reducing the levels of inflammatory immune cells.
Collapse
Affiliation(s)
- Jeonghyeon Moon
- Departments of Immunobiology and Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Keun-Hyung Cho
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - JooYeon Jhun
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - JeongWon Choi
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Hyun-Sik Na
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jeong Su Lee
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Seung Yoon Lee
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jun-Ki Min
- Department of Internal Medicine, and the Clinical Medicine Research Institute of Bucheon St. Mary's Hospital, The Catholic University of Korea, Bucheon si, Gyeonggi-do, Korea
| | - Anan Shetty
- Institute of Medical Sciences, Canterbury Christ Church University, Medway Campus, Chatham, Kent, UK
| | - Sung-Hwan Park
- Department of Internal Medicine, Division of Rheumatology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222, Banpo‑daero, Seocho‑gu, Seoul, 06591, Korea
| | - Seok Jung Kim
- Department of Orthopaedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Cheonbo-ro, Uijeongbu-si, Gyeonggi-do, 271, Korea
| | - Mi-La Cho
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
28
|
Zhang Z, Zhang N, Li M, Ma X, Qiu Y. Sappanone a alleviates osteoarthritis progression by inhibiting chondrocyte ferroptosis via activating the SIRT1/Nrf2 signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8759-8770. [PMID: 38832987 DOI: 10.1007/s00210-024-03179-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that cause pain and disability in adults. Chondrocyte ferroptosis is found to be involved in OA progression. Sappanone A has been found as an anti-inflammatory and antioxidative agent in several diseases. This study aims to investigate the effects of sappanone A on OA progression and chondrocyte ferroptosis. IL-1β-induced chondrocytes and destabilization of the medial meniscus (DMM)-induced rats were respectively used as the OA model in vitro and in vivo. The effects of sappanone A on inflammation, extracellular matrix (ECM) metabolism, and ferroptosis were determined. Our results showed that in IL-1β-induced chondrocytes, sappanone A suppressed the production of NO, PGE2, TNF-α, IL-6, iNOS, and COX2. Sappanone A also inhibited the expression of MMP3, MMP13, and ADAMTS5, while increasing collagen II expression. Moreover, sappanone A alleviated cytotoxicity and decreased the levels of intracellular ROS, lipid ROS, MDA, and iron, while increasing GSH levels. Additionally, sappanone A increased the protein expression of SLC7A11 and GPX4. Administration of ferroptosis activator reversed the inhibitory effects of sappanone A on IL-1β-induced inflammation and ECM degradation. More importantly, Sappanone A activated the Nrf2 signaling by targeting SIRT1. The inhibition of sappanone A on ferroptosis was greatly eliminated due to the addition of SIRT1 inhibitor. Furthermore, intra-articular injection of sappanone A mitigated cartilage destruction and ferroptosis in DMM-induced OA rats. In conclusion, sappanone A protects against inflammation and ECM degradation in OA via decreasing chondrocyte ferroptosis by activating the SIRT1/Nrf2 signaling. These findings deepen our understanding of chondrocyte ferroptosis in OA and highlight the therapeutic potential of sappanone A for OA.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Nanzhi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Meng Li
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Xing Ma
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yusheng Qiu
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
29
|
Joung JY, Lee YH, Son CG. An evolutionary perspective for integrating mechanisms of acupuncture therapy. Explore (NY) 2024; 20:103060. [PMID: 39278099 DOI: 10.1016/j.explore.2024.103060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
This study applies an evolutionary medicine perspective to comprehend the therapeutic effects of acupuncture. It draws upon modern evolutionary theory to integrate the currently fragmented theories regarding the efficacy of acupuncture in alleviating pain and promoting healing. We explore the interaction between the nervous and immune systems in the context of survival and homeostasis, and elucidate both the local and systemic effects of acupuncture therapy on pain relief and tissue healing. The mechanisms involved are categorized into two main types: local effects, which include immune cell migration, local vasodilation, and the release of adenosine; and distal systemic effects, which involve the regulation of the descending pain control system and the autonomic nervous system, with a particular focus on the parasympathetic nervous system. In conclusion, this integrated perspective not only deepens our understanding of acupuncture within a scientific narrative but also underscores the need for further research to validate and expand our knowledge, thereby enhancing its scientific credibility and clinical applicability.
Collapse
Affiliation(s)
- Jin-Yong Joung
- Department of Internal Medicine, Daejeon Good-morning Oriental Hospital, 21, Dunsan-ro 123beon-gil, Seo-gu, Daejeon 35240, Republic of Korea; Department of Korean Medicine, Korean Medical College of Daejeon University, Daehak-ro 62, Dong-gu, Daejeon 34520, Republic of Korea
| | - Young Ho Lee
- Department of Anatomy, College of Medicine, Chungnam National University, 266, Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Chang-Gue Son
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Seo-gu, Daejeon 35235, Republic of Korea; Department of Korean Medicine, Korean Medical College of Daejeon University, Daehak-ro 62, Dong-gu, Daejeon 34520, Republic of Korea.
| |
Collapse
|
30
|
Leng T, Zhang L, Ma J, Qu X, Lei B. Intrinsically bioactive multifunctional Poly(citrate-curcumin) for rapid lung injury and MRSA infection therapy. Bioact Mater 2024; 41:158-173. [PMID: 39131630 PMCID: PMC11314446 DOI: 10.1016/j.bioactmat.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
Dysregulated inflammation after trauma or infection could result in the further disease and delayed tissue reconstruction. The conventional anti-inflammatory drug treatment suffers to the poor bioavailability and side effects. Herein, we developed an amphiphilic multifunctional poly (citrate-polyglycol-curcumin) (PCGC) nano oligomer with the robust anti-inflammatory activity for treating acute lung injury (ALI) and Methicillin-resistant staphylococcus aureus (MRSA) infected wound. PCGC demonstrated the sustained curcumin release, inherent photoluminescence, good cellular compatibility, hemocompatibility, robust antioxidant activity and enhanced cellular uptake. PCGC could efficiently scavenge nitrogen-based free radicals, oxygen-based free radicals, and intracellular oxygen species, enhance the endothelial cell migration and reduce the expression of pro-inflammatory factors through the NF-κB signal pathway. Combined the anti-inflammation and antioxidant properties, PCGC can shortened the inflammatory process. In animal model of ALI, PCGC was able to reduce the pulmonary edema, bronchial cell infiltration, and lung inflammation, while exhibiting rapid metabolic behavior in vivo. The MRSA-infection wound model showed that PCGC significantly reduced the expression of pro-inflammatory factors, promoted the angiogenesis and accelerated the wound healing. The transcriptome sequencing and molecular mechanism studies further demonstrated that PCGC could inhibit multiple inflammatory related pathways including TNFAIP3, IL-15RA, NF-κB. This work demonstrates that PCGC is efficient in resolving inflammation and promotes the prospect of application in inflammatory diseases as the drug-loaded therapeutic system.
Collapse
Affiliation(s)
- Tongtong Leng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Long Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Junping Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Xiaoyan Qu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Bo Lei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| |
Collapse
|
31
|
Wang L, Fu X, Xia T, Yang Z, Zhao R. Myrislignan ameliorates the progression of osteoarthritis: An in vitro and in vivo study. Int Immunopharmacol 2024; 140:112887. [PMID: 39116493 DOI: 10.1016/j.intimp.2024.112887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/19/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Osteoarthritis (OA) is a prevalent disease of the musculoskeletal system that causes functional deterioration and diminished quality of life. Myrislignan (MRL) has a wide range of pharmacological characteristics, including an anti-inflammatory ability. Although inflammation is a major cause of OA, the role of MRL in OA treatment is still not well-understood. In this study, we analyze the anti-inflammatory and anti-ECM degradation effects of MRL both in vivo and in vitro. Rat primary chondrocytes were treated with interleukin-1β (IL-1β) to simulate inflammatory environmental conditions and OA in vitro. The in vivo OA rat model was established by anterior cruciate ligament transection (ACLT) on rat. Our investigation discovered that MRL lowers the IL-1β-activated tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX2) and inducible nitric-oxide synthase (iNOS) expression in chondrocytes. Moreover, MRL effectively alleviates IL-1β-induced extracellular matrix (ECM) degradation and promotes ECM synthesis in chondrocytes by upregulating the mRNA level expression of collagen-II and aggrecan (ACAN), downregulating the expression of matrix metalloproteinases-3,-13 (MMP-3, MMP-13), and a disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5). Gene expression profiles of different groups identified DEGs that were mainly enriched in functions associated with NF-κB signaling pathway, and other highly enriched in functions related to TNF, IL-17, Rheumatoid arthritis and cytokine-cytokine receptor signaling pathways. Venn interaction of DEGs from the abovementioned five pathways showed that Nfkbia, Il1b, Il6, Nfkb1, Ccl2, Mmp3 were highly enriched DEGs. In addition, our research revealed that MRL suppresses NF-κB and modulates the Nrf2/HO-1/JNK signaling pathway activated by IL-1β in chondrocytes. In vivo research shows that MRL slows the progression of OA in rats. Our findings imply that MRL might be a viable OA therapeutic choice.
Collapse
Affiliation(s)
- Liang Wang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Soochow University, Suzhou 215028, Jiangsu, China
| | - Xuejie Fu
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215153, Jiangsu, China
| | - Tingting Xia
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215153, Jiangsu, China
| | - Zhao Yang
- Department of Respiratory Medicine, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215153, Jiangsu, China.
| | - Runze Zhao
- Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital of Soochow University, Suzhou 215028, Jiangsu, China.
| |
Collapse
|
32
|
Li B, Shen Y, Liu S, Yuan H, Liu M, Li H, Zhang T, Du S, Liu X. Identification of immune microenvironment subtypes and clinical risk biomarkers for osteoarthritis based on a machine learning model. Front Mol Biosci 2024; 11:1376793. [PMID: 39484639 PMCID: PMC11524973 DOI: 10.3389/fmolb.2024.1376793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024] Open
Abstract
Background Osteoarthritis (OA) is a degenerative disease with a high incidence worldwide. Most affected patients do not exhibit obvious discomfort symptoms or imaging findings until OA progresses, leading to irreversible destruction of articular cartilage and bone. Therefore, developing new diagnostic biomarkers that can reflect articular cartilage injury is crucial for the early diagnosis of OA. This study aims to explore biomarkers related to the immune microenvironment of OA, providing a new research direction for the early diagnosis and identification of risk factors for OA. Methods We screened and downloaded relevant data from the Gene Expression Omnibus (GEO) database, and the immune microenvironment-related genes (Imr-DEGs) were identified using the ImmPort data set by combining weighted coexpression analysis (WGCNA). Functional enrichment of GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) were conducted to explore the correlation of Imr-DEGs. A random forest machine learning model was constructed to analyze the characteristic genes of OA, and the diagnostic significance was determined by the Receiver Operating Characteristic Curve (ROC) curve, with external datasets used to verify the diagnostic ability. Different immune subtypes of OA were identified by unsupervised clustering, and the function of these subtypes was analyzed by gene set enrichment analysis (GSVA). The Drug-Gene Interaction Database was used to explore the relationship between characteristic genes and drugs. Results Single sample gene set enrichment analysis (ssGSEA) revealed that 16 of 28 immune cell subsets in the dataset significantly differed between OA and normal groups. There were 26 Imr-DEGs identified by WGCNA, showing that functional enrichment was related to immune response. Using the random forest machine learning model algorithm, nine characteristic genes were obtained: BLNK (AUC = 0.809), CCL18 (AUC = 0.692), CD74 (AUC = 0.794), CSF1R (AUC = 0.835), RAC2 (AUC = 0.792), INSR (AUC = 0.765), IL11 (AUC = 0.662), IL18 (AUC = 0.699), and TLR7 (AUC = 0.807). A nomogram was constructed to predict the occurrence and development of OA, and the calibration curve confirmed the accuracy of these 9 genes in OA diagnosis. Conclusion This study identified characteristic genes related to the immune microenvironment in OA, providing new insight into the risk factors of OA.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xinwei Liu
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
33
|
Selim AM, Elsabagh YA, El-Sawalhi MM, Ismail NA, Senousy MA. Serum lncRNA ITGB2-AS1 and ICAM-1 as novel biomarkers for rheumatoid arthritis and osteoarthritis diagnosis. BMC Med Genomics 2024; 17:247. [PMID: 39379962 PMCID: PMC11462822 DOI: 10.1186/s12920-024-01993-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/19/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND The complete circulating long non-coding RNAs (lncRNAs) signature of rheumatoid arthritis (RA) and osteoarthritis (OA) is still uncovered. The lncRNA integrin subunit beta 2 (ITGB2)-anti-sense RNA 1 (ITGB2-AS1) affects ITGB2 expression; however, there is a gap in knowledge regarding its expression and clinical usefulness in RA and OA. This study investigated the potential of serum ITGB2-AS1 as a novel diagnostic biomarker and its correlation with ITGB2 expression and its ligand intercellular adhesion molecule-1 (ICAM-1), disease activity, and severity in RA and primary knee OA patients. SUBJECTS Forty-three RA patients, 35 knee OA patients, and 22 healthy volunteers were included. RESULTS Compared with healthy controls, serum ITGB2-AS1 expression was upregulated in RA patients but wasn't significantly altered in knee OA patients, whereas serum ICAM-1 protein levels were elevated in both diseases. ITGB2-AS1 showed discriminative potential for RA versus controls (AUC = 0.772), while ICAM-1 displayed diagnostic potential for both RA and knee OA versus controls (AUC = 0.804, 0.914, respectively) in receiver-operating characteristic analysis. In the multivariate analysis, serum ITGB2-AS1 and ICAM-1 were associated with the risk of developing RA, while only ICAM-1 was associated with the risk of developing knee OA. A panel combining ITGB2-AS1 and ICAM-1 showed profound diagnostic power for RA (AUC = 0.9, sensitivity = 86.05%, and specificity = 91.67%). Interestingly, serum ITGB2-AS1 positively correlated with disease activity (DAS28) in RA patients and with ITGB2 mRNA expression in both diseases, while ICAM-1 positively correlated with ITGB2 expression in knee OA patients. CONCLUSION Our study portrays serum ITGB2-AS1 as a novel potential diagnostic biomarker of RA that correlates with disease activity. A predictive panel combining ITGB2-AS1 and ICAM-1 could have clinical utility in RA diagnosis. We also spotlight the association of ICAM-1 with knee OA diagnosis. The correlation of serum ITGB2-AS1 with ITGB2 expression in both diseases may be insightful for further mechanistic studies.
Collapse
Affiliation(s)
- Aliaa M Selim
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, 23 Kasr El-Ainy Street, Cairo, 11562, Egypt.
| | - Yumn A Elsabagh
- Department of Rheumatology and Clinical Immunology, Internal Medicine, Kasr Al-Ainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha M El-Sawalhi
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, 23 Kasr El-Ainy Street, Cairo, 11562, Egypt.
| | - Nabila A Ismail
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, 23 Kasr El-Ainy Street, Cairo, 11562, Egypt
| | - Mahmoud A Senousy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, 23 Kasr El-Ainy Street, Cairo, 11562, Egypt
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt
| |
Collapse
|
34
|
Mobasheri A, Rannou F, Ivanavicius S, Conaghan PG. Targeting the TRPV1 pain pathway in osteoarthritis of the knee. Expert Opin Ther Targets 2024; 28:843-856. [PMID: 39450875 DOI: 10.1080/14728222.2024.2416961] [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] [Accepted: 10/11/2024] [Indexed: 10/26/2024]
Abstract
INTRODUCTION The growing prevalence and lack of effective pain therapies for knee osteoarthritis (KOA) results in a substantial unmet need for novel analgesic therapies. The transient receptor potential vanilloid 1 (TRPV1) receptor is expressed in subsets of nociceptive sensory neurons and has major roles in pain transmission and regulation. In the structures of the knee joint, nociceptors are present in abundance. AREAS COVERED TRPV1-expressing nociceptors in the knee represent a rational target to modulate activity at the origin of the pain pathway in KOA and may avoid systemic side effects seen with currently available analgesics. TRPV1 antagonists can induce analgesia, but hyperthermia and thermal hypesthesia side effects have limited their utility. Clinical development of TRPV1 agonists for pain management has progressed further than that of TRPV1 antagonists. Capsaicin and resiniferatoxin have provided proof-of-concept for the modulation of TRPV1 activity in KOA. EXPERT OPINION Intra-articular administration of TRPV1 agonists enables direct delivery to target nerve terminals in the knee, offering a potentially transformative approach for the management of pain associated with KOA. Here, we explore the advances in understanding innervation of the knee joint in KOA, the role of TRPV1-expressing neurons and progress in developing TRPV1 modulators for KOA.
Collapse
Affiliation(s)
- Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Université de Liège, Liège, Belgium
| | - François Rannou
- UFR de Médecine, Faculté de Santé, Université Paris Cité, Paris, France
- Service de Rééducation et de Réadaptation de l'Appareil Locomoteur et des Pathologies du Rachis, Hôpital Cochin, AP-HP, Centre-Université Paris Cité, Paris, France
- INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs (T3S), Campus Saint-Germain-des-Prés, Paris, France
| | | | - Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds & NIHR Leeds Biomedical Research Centre, Leeds, UK
| |
Collapse
|
35
|
Asadikorayem M, Surman F, Weber P, Weber D, Zenobi-Wong M. Zwitterionic Granular Hydrogel for Cartilage Tissue Engineering. Adv Healthc Mater 2024; 13:e2301831. [PMID: 37501337 DOI: 10.1002/adhm.202301831] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Zwitterionic hydrogels have high potential for cartilage tissue engineering due to their ultra-hydrophilicity, nonimmunogenicity, and superior antifouling properties. However, their application in this field has been limited so far, due to the lack of injectable zwitterionic hydrogels that allow for encapsulation of cells in a biocompatible manner. Herein, a novel strategy is developed to engineer cartilage employing zwitterionic granular hydrogels that are injectable, self-healing, in situ crosslinkable and allow for direct encapsulation of cells with biocompatibility. The granular hydrogel is produced by mechanical fragmentation of bulk photocrosslinked hydrogels made of zwitterionic carboxybetaine acrylamide (CBAA), or a mixture of CBAA and zwitterionic sulfobetaine methacrylate (SBMA). The produced microgels are enzymatically crosslinkable using horseradish peroxidase, to quickly stabilize the construct, resulting in a microporous hydrogel. Encapsulated human primary chondrocytes are highly viable and able to proliferate, migrate, and produce cartilaginous extracellular matrix (ECM) in the zwitterionic granular hydrogel. It is also shown that by increasing hydrogel porosity and incorporation of SBMA, cell proliferation and ECM secretion are further improved. This strategy is a simple and scalable method, which has high potential for expanding the versatility and application of zwitterionic hydrogels for diverse tissue engineering applications.
Collapse
Affiliation(s)
- Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - František Surman
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Daniel Weber
- Division of Hand Surgery, University Children's Hospital, Zürich, 8032, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| |
Collapse
|
36
|
Xu L, Ma J, Zhou C, Shen Z, Zhu K, Wu X, Chen Y, Chen T, Lin X. Identification of key hub genes in knee osteoarthritis through integrated bioinformatics analysis. Sci Rep 2024; 14:22437. [PMID: 39341952 PMCID: PMC11439059 DOI: 10.1038/s41598-024-73188-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Knee osteoarthritis (KOA) is a common chronic joint disease globally. Synovial inflammation plays a pivotal role in its pathogenesis, preceding cartilage damage. Identifying biomarkers in osteoarthritic synovial tissues holds promise for early diagnosis and targeted interventions. Gene expression profiles were obtained from the Gene Expression Omnibus database. Subsequent analyses included differential expression gene (DEG) analysis and weighted gene co-expression network analysis (WGCNA) on the combined datasets. We performed functional enrichment analysis on the overlapping genes between DEGs and module genes and constructed a protein-protein interaction network. Using Cytoscape software, we identified hub genes related to the disease and conducted gene set enrichment analysis on these hub genes. The CIBERSORT algorithm was employed to evaluate the correlation between hub genes and the abundance of immune cells within tissues. Finally, Mendelian randomization analysis was utilized to assess the potential of these hub genes as biomarkers. We identified 46 differentially expressed genes (DEGs), comprising 20 upregulated and 26 downregulated genes. Using WGCNA, we constructed a gene co-expression network and selected the most relevant modules, resulting in 24 intersecting genes with the DEGs. KEGG enrichment analysis of the intersecting genes identified the IL-17 signaling pathway, associated with inflammation, as the most significant pathway. Cytoscape software was utilized to rank the candidate genes, with JUN, ATF3, FOSB, NR4A2, and IL6 emerging as the top five based on the Degree algorithm. A nomogram model incorporating these five genes, supported by ROC curve analysis, validated their diagnostic efficacy. Immune infiltration and correlation analysis revealed that macrophages were significantly associated with JUN (p < 0.01), FOSB (p < 0.01), and NR4A2 (p < 0.05). Additionally, T follicular helper cells showed significant associations with ATF3 (p < 0.05), FOSB (p < 0.05), and JUN (p < 0.05). Mendelian randomization analysis provided strong evidence linking JUN (IVW: OR = 0.910, p = 0.005) and IL6 (IVW: OR = 1.024, p = 0.026) with KOA. Through the utilization of various bioinformatics analysis methods, we have pinpointed key hub genes relevant to knee osteoarthritis. These findings hold promise for advancing pre-symptomatic diagnostic strategies and enhancing our understanding of the biological underpinnings behind knee osteoarthritis susceptibility genes.
Collapse
Affiliation(s)
- Lilei Xu
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaqi Ma
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuanlong Zhou
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Acupuncture, Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhe Shen
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kean Zhu
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuewen Wu
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yang Chen
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Chen
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianming Lin
- Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.
- Department of Acupuncture, Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
37
|
Wen Z, Qiu L, Ye Z, Tan X, Xu X, Lu M, Kuang G. The role of Th/Treg immune cells in osteoarthritis. Front Immunol 2024; 15:1393418. [PMID: 39364408 PMCID: PMC11446774 DOI: 10.3389/fimmu.2024.1393418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent clinical condition affecting the entire joint, characterized by its multifactorial etiology and complex pathophysiology. The onset of OA is linked to inflammatory mediators produced by the synovium, cartilage, and subchondral bone, all of which are closely tied to cartilage degradation. Consequently, OA may also be viewed as a systemic inflammatory disorder. Emerging studies have underscored the significance of T cells in the development of OA. Notably, imbalances in Th1/Th2 and Th17/Treg immune cells may play a crucial role in the pathogenesis of OA. This review aims to compile recent advancements in understanding the role of T cells and their Th/Treg subsets in OA, examines the immune alterations and contributions of Th/Treg cells to OA progression, and proposes novel directions for future research, including potential therapeutic strategies for OA.
Collapse
Affiliation(s)
- Zhi Wen
- Department of Joint Orthopedics, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Graduate School of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Liguo Qiu
- Graduate School of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Zifeng Ye
- Graduate School of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Xuyi Tan
- Department of Joint Orthopedics, The Affiliated Hospital, Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Xiaotong Xu
- Department of Joint Orthopedics, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Min Lu
- Department of Joint Orthopedics, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gaoyan Kuang
- Department of Joint Orthopedics, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
38
|
Yu E, Zhang M, Xi C, Yan J. Identification and experimental validation of key genes in osteoarthritis based on machine learning algorithms and single-cell sequencing analysis. Heliyon 2024; 10:e37047. [PMID: 39286216 PMCID: PMC11402953 DOI: 10.1016/j.heliyon.2024.e37047] [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: 04/11/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Osteoarthritis (OA) is a prevalent cause of disability in older adults. Identifying diagnostic markers for OA is essential for elucidating its mechanisms and facilitating early diagnosis. Methods We analyzed 53 synovial tissue samples (n = 30 for OA, n = 23 for the control group) from two datasets in the Gene Express Omnibus (GEO) database. We identified differentially expressed genes (DEGs) between the groups and applied dimensionality reduction using six machine learning algorithms to pinpoint characteristic genes (key genes). We classified the OA samples into subtypes based on these key genes and explored the differences in biological functions and immune characteristics among subtypes, as well as the roles of the key genes. Additionally, we constructed a protein-protein interaction network to predict small molecules that target these genes. Further, we accessed synovial tissue sample data from the single-cell RNA dataset GSE152805, categorized the cells into various types, and examined variations in gene expression and their correlation with OA progression. Validation of key gene expression was conducted in cellular experiments using the qPCR method. Results Four genes AGMAT, MAP3K8, PER1, and XIST, were identified as characteristic genes of OA. All can independently predict the occurrence of OA. With these genes, the OA samples can be clustered into two subtypes, which showed significant differences in functional pathways and immune infiltration. Eight cell types were obtained by analyzing the single-cell RNA data, with synovial intimal fibroblasts (SIF) accounting for the highest proportion in each sample. The key genes were found over-expressed in SIF and significantly correlated with OA progression and the content of immune cells (ICs). We validated the relative levels of key genes in OA and normal cartilage tissue cells, which showed an expression trend consistency with the bioinformatics result except for XIST. Conclusion Four genes, AGMAT, MAP3K8, PER1, and XIST are closely related to the progression of OA, and play as diagnostic and predictive markers in early OA.
Collapse
Affiliation(s)
- Enming Yu
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingshu Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunyang Xi
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
39
|
Singer J, Knezic N, Layne J, Gohring G, Christiansen J, Rothrauff B, Huard J. Enhancing Cartilage Repair: Surgical Approaches, Orthobiologics, and the Promise of Exosomes. Life (Basel) 2024; 14:1149. [PMID: 39337932 PMCID: PMC11432843 DOI: 10.3390/life14091149] [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: 07/28/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Treating cartilage damage is challenging as its ability for self-regeneration is limited. Left untreated, it can progress to osteoarthritis (OA), a joint disorder characterized by the deterioration of articular cartilage and other joint tissues. Surgical options, such as microfracture and cell/tissue transplantation, have shown promise as techniques to harness the body's endogenous regenerative capabilities to promote cartilage repair. Nonetheless, these techniques have been scrutinized due to reported inconsistencies in long-term outcomes and the tendency for the defects to regenerate as fibrocartilage instead of the smooth hyaline cartilage native to joint surfaces. Orthobiologics are medical therapies that utilize biologically derived substances to augment musculoskeletal healing. These treatments are rising in popularity because of their potential to enhance surgical standards of care. More recent developments in orthobiologics have focused on the role of exosomes in articular cartilage repair. Exosomes are nano-sized extracellular vesicles containing cargo such as proteins, lipids, and nucleic acids, and are known to facilitate intercellular communication, though their regenerative potential still needs to be fully understood. This review aims to demonstrate the advancements in cartilage regeneration, highlight surgical and biological treatment options, and discuss the recent strides in understanding the precise mechanisms of action involved.
Collapse
Affiliation(s)
- Jacob Singer
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Noah Knezic
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jonathan Layne
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Greta Gohring
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jeff Christiansen
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Ben Rothrauff
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Johnny Huard
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| |
Collapse
|
40
|
Wang H, Zhang Z, Cheng X, Hou Z, Wang Y, Liu Z, Gao Y. Machine learning algorithm-based biomarker exploration and validation of mitochondria-related diagnostic genes in osteoarthritis. PeerJ 2024; 12:e17963. [PMID: 39282111 PMCID: PMC11397131 DOI: 10.7717/peerj.17963] [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: 09/19/2023] [Accepted: 08/01/2024] [Indexed: 09/18/2024] Open
Abstract
The role of mitochondria in the pathogenesis of osteoarthritis (OA) is significant. In this study, we aimed to identify diagnostic signature genes associated with OA from a set of mitochondria-related genes (MRGs). First, the gene expression profiles of OA cartilage GSE114007 and GSE57218 were obtained from the Gene Expression Omnibus. And the limma method was used to detect differentially expressed genes (DEGs). Second, the biological functions of the DEGs in OA were investigated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Wayne plots were employed to visualize the differentially expressed mitochondrial genes (MDEGs) in OA. Subsequently, the LASSO and SVM-RFE algorithms were employed to elucidate potential OA signature genes within the set of MDEGs. As a result, GRPEL and MTFP1 were identified as signature genes. Notably, GRPEL1 exhibited low expression levels in OA samples from both experimental and test group datasets, demonstrating high diagnostic efficacy. Furthermore, RT-qPCR analysis confirmed the reduced expression of Grpel1 in an in vitro OA model. Lastly, ssGSEA analysis revealed alterations in the infiltration abundance of several immune cells in OA cartilage tissue, which exhibited correlation with GRPEL1 expression. Altogether, this study has revealed that GRPEL1 functions as a novel and significant diagnostic indicator for OA by employing two machine learning methodologies. Furthermore, these findings provide fresh perspectives on potential targeted therapeutic interventions in the future.
Collapse
Affiliation(s)
- Hongbo Wang
- Department of Urology Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zongye Zhang
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xingbo Cheng
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenxing Hou
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yubo Wang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, Henan, China
| | - Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
41
|
Altahla R, Tao X. Thioredoxin-Interacting Protein's Role in NLRP3 Activation and Osteoarthritis Pathogenesis by Pyroptosis Pathway: In Vivo Study. Metabolites 2024; 14:488. [PMID: 39330495 PMCID: PMC11433649 DOI: 10.3390/metabo14090488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Thioredoxin-interacting protein (TXNIP) has been involved in oxidative stress and activation of the NOD-like receptor protein-3 (NLRP3) inflammasome, directly linking it to the pyroptosis pathway. Furthermore, pyroptosis may contribute to the inflammatory process in osteoarthritis (OA). The purpose of this study was to investigate the role of TXNIP in activating the NLRP3 inflammasome through the pyroptosis pathway in an OA rat model. Destabilization of the medial meniscus (DMM) was induced in the OA model with intra-articular injections of adeno-associated virus (AAV) overexpressing (OE) or knocking down (KD) TXNIP. A total of 48 healthy rats were randomly divided into six groups (N = 8 each). During the experiment, the rats' weights, mechanical pain thresholds, and thermal pain thresholds were measured weekly. Morphology staining, micro-CT, 3D imaging, and immunofluorescence (IF) staining were used to measure the expression level of TXNIP, and ELISA techniques were employed. OE-TXNIP-AAV in DMM rats aggravated cartilage destruction and subchondral bone loss, whereas KD-TXNIP slowed the progression of OA. The histological results showed that DMM modeling and OE-TXNIP-AAV intra-articular injection caused joint structure destruction, decreased anabolic protein expression, and increased catabolic protein expression and pyroptosis markers. Conversely, KD-TXNIP-AAV slowed joint degeneration. OE-TXNIP-AVV worsened OA by accelerating joint degeneration and damage, while KD-TXNIP-AAV treatment had a protective effect.
Collapse
Affiliation(s)
- Ruba Altahla
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xu Tao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
42
|
Ju J, Ma M, Zhang Y, Ding Z, Lin P, Chen J. Transcriptome sequencing reveals inflammation and macrophage heterogeneity in subacromial bursa from degenerative shoulder disorders. Connect Tissue Res 2024; 65:383-396. [PMID: 39109563 DOI: 10.1080/03008207.2024.2386548] [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: 04/22/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 10/17/2024]
Abstract
PURPOSE We aimed to investigate the transcriptomic alterations that occur in the subacromial bursa (SAB) following degenerative or traumatic shoulder diseases. MATERIALS AND METHODS RNA sequencing was employed to evaluate the transcriptomic alterations of the SAB in individuals afflicted with degenerative rotator cuff tear (RCT), traumatic RCT and proximal humerus fracture (PHF). To gain insights into the biological significance of differentially expressed genes (DEGs), we conducted an enrichment analysis utilizing Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. We further utilized single-cell RNA sequencing datasets of SAB from a recently published study to explore the associated cellular dynamics and alterations. RESULTS We detected 1,790 up-regulated and 1,964 down-regulated DEGs between degenerative RCT and PHF, 2,085 up-regulated and 1,919 down-regulated DEGs between degenerative RCT and traumatic RCT, and 20 up-regulated and 12 down-regulated DEGs between traumatic RCT and PHF. Given the similar expression pattern between traumatic RCT and PHF, they were integrated as the traumatic group. In comparison with the traumatic group, 1,983 up-regulated and 2,205 down-regulated DEGs were detected in degenerative SAB. Enrichment analysis of up-regulated DEGs uncovered an elevated inflammatory and immunologic responses in degenerative SAB. Single-cell transcriptomic analysis revealed macrophage represented the immune cell with the most DEGs between the degenerative and traumatic RCT. CONCLUSION Our results revealed that the SAB in degenerative RCT exhibited a different transcriptional signature compared to that in traumatic RCT, and enrichment analysis showed immunologic and inflammatory activations. Macrophages may play a fundamental role in this process.
Collapse
Affiliation(s)
- Jiabao Ju
- Department of Trauma & Orthopedics, Peking University People's Hospital, Beijing, China
| | - Mingtai Ma
- Department of Trauma & Orthopedics, Peking University People's Hospital, Beijing, China
| | - Yichong Zhang
- Department of Trauma & Orthopedics, Peking University People's Hospital, Beijing, China
| | - Zhentao Ding
- Department of Trauma & Orthopedics, Peking University People's Hospital, Beijing, China
| | - Pingping Lin
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China
| | - Jianhai Chen
- Department of Trauma & Orthopedics, Peking University People's Hospital, Beijing, China
| |
Collapse
|
43
|
Peng C, Yang L, Labens R, Gao Y, Zhu Y, Li J. A systematic review and meta-analysis of the efficacy of platelet-rich plasma products for treatment of equine joint disease. Equine Vet J 2024; 56:858-869. [PMID: 38185481 DOI: 10.1111/evj.14042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 11/25/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Equine joint disease including septic arthritis (SA) and osteoarthritis (OA) is a critical cause of equine lameness. Platelet-rich plasma (PRP) is one of the most popular regenerative therapies to treat equine OA, even SA, but the evidence in support of the treatment is conflicting. OBJECTIVES The aim of the study was to systematically review the current evidence on PRP products used for SA and OA, as well as the efficacy of PRP products as treatment for OA on the basis of a meta-analysis of the available literature. STUDY DESIGN Systematic review and meta-analysis. METHODS A systematic search of relevant databases (PubMed, Web of Science, Scopus) was performed to identify studies published from 2013 to 2023, in accordance with the PRISMA guidelines. Randomised controlled trials, non-randomised trials and controlled laboratory studies that used at least one type of PRP products were included. Dichotomous outcomes were presented using odds ratios (ORs) and 95% confidence intervals (95% CIs). RESULTS A total of 21 publications were identified in the systematic review and 5 of them in the meta-analysis. These publications involved various types of PRP products and reported different outcomes. Although most of the studies were associated with a high risk of bias, the overall estimated effect was consistent with a significant improvement in the PRP products treatment group compared with the control group (OR: 15.32; 95% CI: 3.00-78.15; p < 0.05). There was a significant improvement in clinical performance outcomes between the groups (OR: 36.64; 95% CI: 3.69-364.30; p < 0.05). CONCLUSION PRP products as intra-articular treatment are likely efficacious for treatment of equine OA and have potential for treating SA. These conclusions might be affected by the limited number of randomised controlled studies and high variability of different types of PRP products. To better evaluate the efficacy of PRP, a widely recognised classification system and the utilisation of randomised, blinded, equivalency or non-inferiority trials are required.
Collapse
Affiliation(s)
- Cong Peng
- Equine Clinical Diagnostic Center, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Luo Yang
- Equine Clinical Diagnostic Center, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Raphael Labens
- School of Agricultural, Environmental and Veterinary Sciences, Charles Sturt University, Bathurst, New South Wales, Australia
| | - Yu Gao
- University of Veterinary Medicine Hannover, Hanover, Lower Saxony, Germany
| | - Yiping Zhu
- Equine Clinical Diagnostic Center, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jing Li
- Equine Clinical Diagnostic Center, College of Veterinary Medicine, China Agricultural University, Beijing, China
- China Agricultural University Veterinary Teaching Hospital, Beijing, China
| |
Collapse
|
44
|
Ye X, Shen Z, Li X, Zhang B, Shen G, Wu L. Microfragmented adipose tissue versus platelet-rich plasma in the treatment of knee osteoarthritis: a systematic review and meta-analysis. Acta Orthop Belg 2024; 90:549-558. [PMID: 39851028 DOI: 10.52628/90.3.12669] [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: 01/25/2025]
Abstract
This meta-analysis focuses on the controversial efficacy and safety of microfragmented adipose tissue (MFAT) as compared with platelet-rich plasma (PRP) in the clinical treatment of knee osteoarthritis (KOA). We have attempted to provide an evidence-based medicine protocol for the conservative treatment of KOA. Researchers collected and compared randomized controlled trials (RCTs) that used microfragmented adipose tissue and platelet-rich plasma to treat knee osteoarthritis. We searched CNKI, Wanfang Database, CMJD, PubMed, Sinomed, Cochrane Library, and Embase for studies published up to May 31, 2023. Two investigators independently screened literature, extracted data, and assessed bias risk using the Cochrane bias risk tool. The researchers then performed a meta-analysis using Revman 5.4 statistics software provided by the Cochrane Library. A total of 4 randomized controlled trials involving 266 patients (326 knees) were included. There were 161 knees in the MFAT group and 165 knees in the PRP group. Meta-analysis showed a statistically significant difference in VAS scores between the MFAT group and the PRP group at 12 months after treatment [MD=0.99, 95% CI (0.31, 1.67), P=0.004]. This result showed that VAS scores were lower in the PRP group than in the MFAT group, and that PRP injection reduced pain more effectively than MFAT injection. At 6 months after treatment, Tegner activity scale scores in the MFAT group were higher than that in the PRP group [MD=0.65, 95% CI (0.11, 1.19), P=0.02], and the difference was statistically significant. There were no significant differences in the remaining indicators between the two groups. Based on this meta-analysis, PRP appears to be more effective than MFAT in treating KOA in terms of long-term pain relief. However, MFAT was superior to PRP in improving short-term activity function. Overall, there was no significant difference between MFAT and PRP in the treatment of KOA. In addition, MFAT does not increase the risk of adverse events compared to PRP. However, at present, there are few clinical studies on MFAT and PRP, which need to be verified by more rigorously designed clinical trials.
Collapse
|
45
|
Ayala S, Matan SO, Delco ML, Fortier LA, Cohen I, Bonassar LJ. Degradation of lubricating molecules in synovial fluid alters chondrocyte sensitivity to shear strain. J Orthop Res 2024. [PMID: 39182184 DOI: 10.1002/jor.25960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/01/2024] [Accepted: 08/03/2024] [Indexed: 08/27/2024]
Abstract
Articular joints facilitate motion and transfer loads to underlying bone through a combination of cartilage tissue and synovial fluid, which together generate a low-friction contact surface. Traumatic injury delivered to cartilage and the surrounding joint capsule causes secretion of proinflammatory cytokines by chondrocytes and the synovium, triggering cartilage matrix breakdown and impairing the ability of synovial fluid to lubricate the joint. Once these inflammatory processes become chronic, posttraumatic osteoarthritis (PTOA) development begins. However, the exact mechanism by which negative alterations to synovial fluid leads to PTOA pathogenesis is not fully understood. We hypothesize that removing the lubricating macromolecules from synovial fluid alters the relationship between mechanical loads and subsequent chondrocyte behavior in injured cartilage. To test this hypothesis, we utilized an ex vivo model of PTOA that involves subjecting cartilage explants to a single rapid impact followed by continuous articulation within a lubricating bath of either healthy synovial fluid, phosphate-buffered saline (PBS), synovial fluid treated with hyaluronidase, or synovial fluid treated with trypsin. These treatments degrade the main macromolecules attributed with providing synovial fluid with its lubricating properties; hyaluronic acid and lubricin. Explants were then bisected and fluorescently stained to assess global and depth-dependent cell death, caspase activity, and mitochondrial depolarization. Explants were tested via confocal elastography to determine the local shear strain profile generated in each lubricant. These results show that degrading hyaluronic acid or lubricin in synovial fluid significantly increases middle zone chondrocyte damage and shear strain loading magnitudes, while also altering chondrocyte sensitivity to loading.
Collapse
Affiliation(s)
- Steven Ayala
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Salman O Matan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Michelle L Delco
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Lisa A Fortier
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Itai Cohen
- Department of Physics, Cornell University, Ithaca, New York, USA
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
46
|
Sun Y, Yang H, Guo J, Du J, Han S, Yang X. Identification of HTRA1, DPT and MXRA5 as potential biomarkers associated with osteoarthritis progression and immune infiltration. BMC Musculoskelet Disord 2024; 25:647. [PMID: 39148085 PMCID: PMC11325630 DOI: 10.1186/s12891-024-07758-7] [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: 12/05/2023] [Accepted: 08/05/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Our study aimed to identify potential specific biomarkers for osteoarthritis (OA) and assess their relationship with immune infiltration. METHODS We utilized data from GSE117999, GSE51588, and GSE57218 as training sets, while GSE114007 served as a validation set, all obtained from the GEO database. First, weighted gene co-expression network analysis (WGCNA) and functional enrichment analysis were performed to identify hub modules and potential functions of genes. We subsequently screened for potential OA biomarkers within the differentially expressed genes (DEGs) of the hub module using machine learning methods. The diagnostic accuracy of the candidate genes was validated. Additionally, single gene analysis and ssGSEA was performed. Then, we explored the relationship between biomarkers and immune cells. Lastly, we employed RT-PCR to validate our results. RESULTS WGCNA results suggested that the blue module was the most associated with OA and was functionally associated with extracellular matrix (ECM)-related terms. Our analysis identified ALB, HTRA1, DPT, MXRA5, CILP, MPO, and PLAT as potential biomarkers. Notably, HTRA1, DPT, and MXRA5 consistently exhibited increased expression in OA across both training and validation cohorts, demonstrating robust diagnostic potential. The ssGSEA results revealed that abnormal infiltration of DCs, NK cells, Tfh, Th2, and Treg cells might contribute to OA progression. HTRA1, DPT, and MXRA5 showed significant correlation with immune cell infiltration. The RT-PCR results also confirmed these findings. CONCLUSIONS HTRA1, DPT, and MXRA5 are promising biomarkers for OA. Their overexpression strongly correlates with OA progression and immune cell infiltration.
Collapse
Affiliation(s)
- Yunchao Sun
- Hebei North University, Zhangjiakou, Hebei, 075000, China
- Department of orthopaedic surgery, Huabeiyiliao Jiankangjituan Fengfeng Zongyiyuan, Handan, Hebei, 056000, China
| | - Hui Yang
- Department of orthopaedic surgery, Huabeiyiliao Jiankangjituan Fengfeng Zongyiyuan, Handan, Hebei, 056000, China
| | - Jiaquan Guo
- Department of orthopaedic surgery, Huabeiyiliao Jiankangjituan Fengfeng Zongyiyuan, Handan, Hebei, 056000, China
| | - Jian Du
- Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Shoujiang Han
- Department of orthopaedic surgery, Huabeiyiliao Jiankangjituan Fengfeng Zongyiyuan, Handan, Hebei, 056000, China.
| | - Xinming Yang
- Hebei North University, Zhangjiakou, Hebei, 075000, China.
- Department of orthopaedic surgery, The first affiliated hospital of Hebei North University, Zhangjiakou, Hebei, 075000, China.
| |
Collapse
|
47
|
Huang H, Yang L, He H, Zhou B, Qin Z, Zheng L, Shen C. Construction of mitochondrial-targeting nano-prodrug for enhanced Rhein delivery and treatment for osteoarthritis in vitro. Int J Pharm 2024; 661:124397. [PMID: 38945463 DOI: 10.1016/j.ijpharm.2024.124397] [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: 04/21/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Rhein, a natural anthraquinone compound derived from traditional Chinese medicine, exhibits potent anti-inflammatory properties via modulating the level of Reactive oxygen or nitrogen species (RONS). Nevertheless, its limited solubility in water, brief duration of plasma presence, as well as its significant systemic toxicity, pose obstacles to its in vivo usage, necessitating the creation of a reliable drug delivery platform to circumvent these difficulties. In this study, an esterase-responsive and mitochondria-targeted nano-prodrug was synthesized by conjugating Rhein with the polyethylene glycol (PEG)-modified triphenyl phosphonium (TPP) molecule, forming TPP-PEG-RH, which could spontaneously self-assemble into RPT NPs when dispersed in aqueous media. The TPP outer layer of these nanoparticles enhances their pharmacokinetic profile, facilitates efficient delivery to mitochondria, and promotes cellular uptake, thereby enabling enhanced accumulation in mitochondria and improved therapeutic effects in vitro. The decline in RONS was observed in IL-1β-stimulated chondrocyte after RPT NPs treating. RPT NPs also exert excellent anti-inflammatory (IL-1β, TNF-α, IL-6 and MMP-13) and antioxidative effects (Cat and Sod) via the Nrf2 signalling pathway, upregulation of cartilage related genes (Col2a1 and Acan). Moreover, RPT NPs shows protection of mitochondrial membrane potential and inhibition of chondrocyte apoptosis. Moreover, These findings suggest that the mitochondria-targeted polymer-Rhein conjugate may offer a therapeutic solution for patients suffering from chronic joint disorders, by attenuating the progression of osteoarthritis (OA).
Collapse
Affiliation(s)
- Hongjun Huang
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, China
| | - Lerong Yang
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, China
| | - Haoqiang He
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Bo Zhou
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zainen Qin
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Chong Shen
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, China.
| |
Collapse
|
48
|
Tu B, Zhu Z, Lu P, Fang R, Peng C, Tong J, Ning R. Proteomic and lipidomic landscape of the infrapatellar fat pad and its clinical significance in knee osteoarthritis. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159513. [PMID: 38788831 DOI: 10.1016/j.bbalip.2024.159513] [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: 06/15/2023] [Revised: 04/25/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Osteoarthritis (OA) is a prevalent joint disease that can be exacerbated by lipid metabolism disorders. The intra-articular fat pad (IFP) has emerged as an active participant in the pathological changes of knee OA (KOA). However, the proteomic and lipidomic differences between IFP tissues from KOA and control individuals remain unclear. Samples of IFP were collected from individuals with and without OA (n = 6, n = 6). Subsequently, these samples underwent liquid chromatography/mass spectrometry-based label-free quantitative proteomic and lipidomic analysis to identify differentially expressed proteins (DEPs) and lipid metabolites (DELMs). The DEPs were further subjected to enrichment analysis, and hub DEPs were identified using multiple algorithms. Additionally, an OA diagnostic model was constructed based on the identified hub DEPs or DELMs. Furthermore, CIBERSORT was utilized to investigate the correlation between hub protein expression and immune-related modules in IFP of OA. Our results revealed the presence of 315 DEPs and eight DELMs in IFP of OA patients compared to the control group. Enrichment analysis of DEPs highlighted potential alterations in pathways related to coagulation, complement, fatty acid metabolism, and adipogenesis. The diagnostic model incorporating four hub DEPs (AUC = 0.861) or eight DELMs (AUC = 0.917) exhibited excellent clinical validity for diagnosing OA. Furthermore, the hub DEPs were found to be associated with immune dysfunction in IFP of OA. This study presents a distinct proteomic and lipidomic landscape of IFP between individuals with OA and those without. These findings provide valuable insights into the molecular changes associated with potential mechanisms underlying OA.
Collapse
Affiliation(s)
- Bizhi Tu
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), 390 Huaihe Road, Hefei 230061, Anhui, China
| | - Zheng Zhu
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), 390 Huaihe Road, Hefei 230061, Anhui, China
| | - Peizhi Lu
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), 390 Huaihe Road, Hefei 230061, Anhui, China; Department of Orthopedics, Bengbu Medical College, Bengbu City 233000, China
| | - Run Fang
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), 390 Huaihe Road, Hefei 230061, Anhui, China
| | - Cheng Peng
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), 390 Huaihe Road, Hefei 230061, Anhui, China
| | - Jun Tong
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), 390 Huaihe Road, Hefei 230061, Anhui, China
| | - Rende Ning
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), 390 Huaihe Road, Hefei 230061, Anhui, China; Department of Orthopedics, Bengbu Medical College, Bengbu City 233000, China.
| |
Collapse
|
49
|
Zhang X, Liu T, Ran C, Wang W, Piao F, Yang J, Tian S, Li L, Zhao D. Immunoregulatory paracrine effect of mesenchymal stem cells and mechanism in the treatment of osteoarthritis. Front Cell Dev Biol 2024; 12:1411507. [PMID: 39129785 PMCID: PMC11310049 DOI: 10.3389/fcell.2024.1411507] [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: 04/03/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease caused by chronic inflammation that damages articular cartilage. At present, the treatment of OA includes drug therapy to relieve symptoms and joint replacement therapy for advanced OA. However, these palliatives cannot truly block the progression of the disease from the immunological pathogenesis of OA. In recent years, bone marrow mesenchymal stem cell (BMSC) transplantation has shown great potential in tissue engineering repair. In addition, many studies have shown that BMSC paracrine signals play an important role in the treatment of OA through immune regulation and suppressing inflammation. At present, the mechanism of inflammation-induced OA and the use of BMSC transplantation in joint repair have been reviewed, but the mechanism and significance of BMSC paracrine signals in the treatment of OA have not been fully reviewed. Therefore, this article focused on the latest research progress on the paracrine effects of BMSCs in the treatment of OA and the related mechanisms by which BMSCs secrete cytokines to inhibit the inflammatory response, regulate immune balance, and promote cell proliferation and differentiation. In addition, the application potential of BMSC-Exos as a new type of cell-free therapy for OA is described. This review aimed to provide systematic theoretical support for the clinical application of BMSC transplantation in the treatment of OA.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Tianhao Liu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Chunxiao Ran
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Weidan Wang
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Fengyuan Piao
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Jiahui Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Simiao Tian
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Lu Li
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Dewei Zhao
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
50
|
Everts PA, Lana JF, Alexander RW, Dallo I, Kon E, Ambach MA, van Zundert A, Podesta L. Profound Properties of Protein-Rich, Platelet-Rich Plasma Matrices as Novel, Multi-Purpose Biological Platforms in Tissue Repair, Regeneration, and Wound Healing. Int J Mol Sci 2024; 25:7914. [PMID: 39063156 PMCID: PMC11277244 DOI: 10.3390/ijms25147914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/07/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Autologous platelet-rich plasma (PRP) preparations are prepared at the point of care. Centrifugation cellular density separation sequesters a fresh unit of blood into three main fractions: a platelet-poor plasma (PPP) fraction, a stratum rich in platelets (platelet concentrate), and variable leukocyte bioformulation and erythrocyte fractions. The employment of autologous platelet concentrates facilitates the biological potential to accelerate and support numerous cellular activities that can lead to tissue repair, tissue regeneration, wound healing, and, ultimately, functional and structural repair. Normally, after PRP preparation, the PPP fraction is discarded. One of the less well-known but equally important features of PPP is that particular growth factors (GFs) are not abundantly present in PRP, as they reside outside of the platelet alpha granules. Precisely, insulin-like growth factor-1 (IGF-1) and hepatocyte growth factor (HGF) are mainly present in the PPP fraction. In addition to their roles as angiogenesis activators, these plasma-based GFs are also known to inhibit inflammation and fibrosis, and they promote keratinocyte migration and support tissue repair and wound healing. Additionally, PPP is known for the presence of exosomes and other macrovesicles, exerting cell-cell communication and cell signaling. Newly developed ultrafiltration technologies incorporate PPP processing methods by eliminating, in a fast and efficient manner, plasma water, cytokines, molecules, and plasma proteins with a molecular mass (weight) less than the pore size of the fibers. Consequently, a viable and viscous protein concentrate of functional total proteins, like fibrinogen, albumin, and alpha-2-macroglobulin is created. Consolidating a small volume of high platelet concentrate with a small volume of highly concentrated protein-rich PPP creates a protein-rich, platelet-rich plasma (PR-PRP) biological preparation. After the activation of proteins, mainly fibrinogen, the PR-PRP matrix retains and facilitates interactions between invading resident cells, like macrophages, fibroblast, and mesenchymal stem cells (MSCs), as well as the embedded concentrated PRP cells and molecules. The administered PR-PRP biologic will ultimately undergo fibrinolysis, leading to a sustained release of concentrated cells and molecules that have been retained in the PR-PRP matrix until the matrix is dissolved. We will discuss the unique biological and tissue reparative and regenerative properties of the PR-PRP matrix.
Collapse
Affiliation(s)
- Peter A. Everts
- Gulf Coast Biologics, A Non-Profit Organization, Fort Myers, FL 33916, USA
- OrthoRegen Group, Max-Planck University, Indaiatuba 13334-170, SP, Brazil;
| | - José Fábio Lana
- OrthoRegen Group, Max-Planck University, Indaiatuba 13334-170, SP, Brazil;
| | - Robert W. Alexander
- Regenevita Biocellular Aesthetic & Reconstructive Surgery, Cranio-Maxillofacial Surgery, Regenerative and Wound Healing, Hamilton, MT 59840, USA;
- Department of Surgery & Maxillofacial Surgery, School of Medicine & Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Ignacio Dallo
- Unit of Biological Therapies and MSK Interventionism, Department of Orthopaedic Surgery and Sports Medicine, Sport Me Medical Center, 41013 Seville, Spain;
| | - Elizaveta Kon
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Mary A. Ambach
- BioEvolve, San Diego Orthobiologics and Sports Center, San Diego, CA 92024, USA
| | - André van Zundert
- Department of Anaesthesia and Perioperative Medicine, Royal Brisbane and Women’s Hospital, Brisbane and The University of Queensland, Brisbane 4072, Australia;
| | - Luga Podesta
- Bluetail Medical Group & Podesta Orthopedic Sports Medicine, Naples, FL 34109, USA;
- Physical Medicine & Rehabilitation Orlando College of Osteopathic Medicine, Orlando, FL 32806, USA
| |
Collapse
|