1
|
Matsuura T, Fukuoka S, Sasaki T, Takaoka K. Advanced Varus Deformity Associated with Medial Knee Osteoarthritis is a Potential Predictor of Anterior Cruciate Ligament Tear and Risk for Suitable Unicompartmental Knee Arthroplasty. J Arthroplasty 2025:S0883-5403(25)00020-8. [PMID: 39824233 DOI: 10.1016/j.arth.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/23/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Previous clinical studies suggest that preserving the anterior cruciate ligament (ACL) is crucial for stable knee motion and long-term longevity of the reconstructed knee. The ACL damage or loss often occurs in advanced medial osteoarthritis (OA). This study aimed to investigate the correlation between ACL damage and varus deformity progression as a risk factor for ACL tears in knee OA. METHODS A retrospective study was conducted on medial knee OA patients who have various degrees of varus deformity treated with unicompartmental knee arthroplasty (UKA; n = 165), where ACLs had no or mild damage. Another group with ACL tears or loss and poor range of motion underwent total knee arthroplasty (TKA; n = 184). Surgeries were performed between November 2016 and March 2023. Preoperative varus angles were measured using the hip-knee-ankle angle (HKA) on standing radiographs. The ACL damage was graded using the Oxford system, and the correlation between preoperative HKA and ACL damage was analyzed. RESULTS A significant correlation was found between preoperative varus angle and ACL damage. Severe damage was more frequent in knees with higher varus angles. The cut-off varus angle for a high risk of ACL tear was identified as 11.3º, with HKA above 15º indicating substantial ACL tears. CONCLUSION Medial knee OA with advanced varus deformity is associated with severe ACL tears. A UKA may be preferable for medial knee OA before the varus angle reaches more than 15º.
Collapse
Affiliation(s)
- Takanori Matsuura
- Department of Orthopedic Surgery, Nishinomiya Watanabe Hospital, Hyogo, Japan
| | - Shinichi Fukuoka
- Department of Orthopedic Surgery, Nishinomiya Watanabe Hospital, Hyogo, Japan.
| | - Takeharu Sasaki
- Department of Orthopedic Surgery, Nishinomiya Watanabe Hospital, Hyogo, Japan
| | - Kunio Takaoka
- Department of Orthopedic Surgery, Nishinomiya Watanabe Hospital, Hyogo, Japan
| |
Collapse
|
2
|
Ossendorff R, Kurth S, Wang S, Jaenisch M, Assaf E, Scheidt S, Welle K, Burger C, Wirtz DC, Strauss AC, Schildberg FA. Comparison of Concentration- and Homology-Dependent Effects of the Proinflammatory Cytokine Interleukin-1β (IL-1β) in a Bovine Chondrocyte Inflammation Model. Cells 2024; 14:30. [PMID: 39791731 PMCID: PMC11719847 DOI: 10.3390/cells14010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025] Open
Abstract
Inflammation models with the proinflammatory cytokine interleukin-1β (IL-1β) are widely used in the in vitro investigation of new therapeutic approaches for osteoarthritis (OA). The aim of this study was to systematically analyze the influence of IL-1β in a 3D chondral pellet culture model. Bovine articular chondrocytes were cultured to passage 3 and then placed in pellet culture. Titration of IL-1β (100-0.1 ng/mL) was performed with both human and bovine recombinant protein in chondrocyte culture for 2 weeks. Gene expression of anabolic (collagen 2, aggrecan, cartilage oligomeric protein (COMP), proteoglycan-4 (PRG-4)), catabolic matrix metallo proteinases (MMP-3, MMP-13), dedifferentiation (collagen 1) markers and inflammatory cytokines IL-6 and IL-8 was determined. Analysis of the cell culture medium was performed for the inflammatory markers IL-6 and nitric oxide (NO). In general, the influence of IL-1β was shown by a decrease in the expression of anabolic markers (collagen 2, aggrecan, PRG-4), whereas the catabolic markers MMP-3 and MMP-13 as well as the inflammatory markers IL-6 and IL-8 were significantly increased. This was observed both at the early time point (day 4) and at the late time point (day 14). The described inflammatory effects were confirmed by increased concentration-dependent release of NO and IL-6. The threshold concentration for a detectable effect compared to control differed between groups, but was reached earlier by homologous application of IL-1β. This study provides a systematic evaluation of IL-1β-specific effects on chondrocytes in a 3D pellet culture model, which is highly relevant for comparisons of studies in OA-specific drug development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
3
|
Zhang Y, Li J, Liu J, Gao Y, Li K, Zhao X, Liu Y, Wang D, Hu X, Wang Z. Ferroptosis in Osteoarthritis: Towards Novel Therapeutic Strategy. Cell Prolif 2024. [DOI: 10.1111/cpr.13779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/09/2024] [Indexed: 01/06/2025] Open
Abstract
ABSTRACTOsteoarthritis (OA) is a chronic, degenerative joint disease primarily characterised by damage to the articular cartilage, synovitis and persistent pain, and has become one of the most common diseases worldwide. In OA cartilage, various forms of cell death have been identified, including apoptosis, necroptosis and autophagic cell death. Ever‐growing observations indicate that ferroptosis, a newly‐discovered iron‐dependent form of regulated cell death, is detrimental to OA occurrence and progression. In this review, we first analyse the pathogenetic mechanisms of OA by which iron overload, inflammatory response and mechanical stress contribute to ferroptosis. We then discuss how ferroptosis exacerbates OA progression, focusing on its impact on chondrocyte viability, synoviocyte populations and extracellular matrix integrity. Finally, we highlight several potential therapeutic strategies targeting ferroptosis that could be explored for the treatment of OA.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Genetics and Cell Biology, School of Basic Medicine Qingdao University Qingdao China
- Department of Reproductive Medicine The Affiliated Hospital of Qingdao University Qingdao China
| | - Jing Li
- Department of Hematology Rizhao People's Hospital Rizhao China
| | - Jiane Liu
- Department of Genetics and Cell Biology, School of Basic Medicine Qingdao University Qingdao China
- Department of Reproductive Medicine The Affiliated Hospital of Qingdao University Qingdao China
| | - Yan Gao
- Department of Hematology The Affiliated Hospital of Qingdao University Qingdao China
| | - Kehan Li
- Department of Genetics and Cell Biology, School of Basic Medicine Qingdao University Qingdao China
| | - Xinyu Zhao
- Department of Genetics and Cell Biology, School of Basic Medicine Qingdao University Qingdao China
| | - Yufeng Liu
- Department of Genetics and Cell Biology, School of Basic Medicine Qingdao University Qingdao China
| | - Daijie Wang
- International Joint Laboratory of Medicinal Food R&D and Health Products Creation/Biological Engineering Technology Innovation Center of Shandong Province Heze Branch of Qilu University of Technology (Shandong Academy of Sciences) Heze China
| | - Xiao Hu
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin Diseases; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
| | - Zheng Wang
- Department of Genetics and Cell Biology, School of Basic Medicine Qingdao University Qingdao China
- Department of Reproductive Medicine The Affiliated Hospital of Qingdao University Qingdao China
| |
Collapse
|
4
|
Tokeshi S, Suzuki-Narita M, Tajiri I, Inage K, Takeuchi J, Arai T, Kawarai Y, Terakawa H, Ohtori S, Orita S. Comparison of effects of intra-articular diclofenac etalhyaluronate and hyaluronic acid in a monoiodoacetate rat osteoarthritis model. J Orthop Res 2024. [PMID: 39528346 DOI: 10.1002/jor.26012] [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: 05/15/2024] [Revised: 08/30/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Diclofenac etalhyaluronate (DF-HA) sustained diclofenac release with the effects of hyaluronic acid (HA), offering long-term analgesia in osteoarthritis. In this study, the effects of DF-HA on pain improvement and osteoarthritis were evaluated in a rat knee monoiodoacetate-induced osteoarthritis model compared to HA. Eight rats per group had been injected with monoiodoacetate (2.0 mg) or saline in the right knee for 4 weeks and were injected with either DF-HA (1.25 mg/kg; 0.5 mg), HA (0.5 mg), vehicle which was a substrate without DF-HA (50 μL), or saline and followed for 4 weeks. Mechanical plantar skin sensitivity was assessed weekly using the von Frey assay. Osteoarthritis changes were monitored with Larsen scores via CT imaging at every 2 weeks. The articular cartilage was analyzed using OARSI scores through H&E, Safranin-O staining at 8 weeks. The percentage of Iba-1 positive microglia in the spinal dorsal horn and of FG + CGRP-labeled cells among FG-positive cells in the dorsal root ganglion were evaluated by immunohistochemical staining. TNF-α and IL-6 mRNA expression levels in the knee synovium were evaluated by PCR. The DF-HA showed significantly improved pain hypersensitivity compared with the HA at 6-8 weeks. The percentage of Iba-1-positive microglia was significantly lower than that in the vehicle and the percentage of FG + CGRP/FG was significantly lower than that in the HA. OARSI scores did not differ among treatment groups, Larsen scores indicated lower in the DF-HA than in the vehicle. DF-HA was as effective as HA in joint protection and significantly improved inflammatory pain compared to HA.
Collapse
Affiliation(s)
- Soichiro Tokeshi
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chuo-ku, Japan
| | - Miyako Suzuki-Narita
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chuo‑ku, Japan
| | - Ikuko Tajiri
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chuo-ku, Japan
| | - Kazuhide Inage
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chuo-ku, Japan
| | - Jun Takeuchi
- Medical Affairs, Seikagaku Corporation, Chiyoda-ku, Japan
| | - Takahito Arai
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chuo-ku, Japan
| | - Yuya Kawarai
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chuo-ku, Japan
| | - Hiroakira Terakawa
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chuo-ku, Japan
| | - Seiji Ohtori
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chuo-ku, Japan
| | - Sumihisa Orita
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chuo-ku, Japan
- Center for Frontier Medical Engineering, Chiba University, Inage‑ku, Japan
| |
Collapse
|
5
|
Ohtsuki T, Sato I, Takashita R, Kodama S, Ikemura K, Opoku G, Watanabe S, Furumatsu T, Yamada H, Ando M, Akiyoshi K, Nishida K, Hirohata S. Distribution and Incorporation of Extracellular Vesicles into Chondrocytes and Synoviocytes. Int J Mol Sci 2024; 25:11942. [PMID: 39596012 PMCID: PMC11593503 DOI: 10.3390/ijms252211942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/25/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic disease affecting over 500 million people worldwide. As the population ages and obesity rates rise, the societal burden of OA is increasing. Pro-inflammatory cytokines, particularly interleukin-1β, are implicated in the pathogenesis of OA. Recent studies suggest that crosstalk between cartilage and synovium contributes to OA development, but the mechanisms remain unclear. Extracellular vesicles (EVs) were purified from cell culture-conditioned medium via ultracentrifugation and confirmed using transmission electron microscopy, nanoparticle tracking analysis, and western blotting. We demonstrated that EVs were taken up by human synoviocytes and chondrocytes in vitro, while in vivo experiments revealed that fluorescent-labelled EVs injected into mouse joints were incorporated into chondrocytes and synoviocytes. EV uptake was significantly inhibited by dynamin-mediated endocytosis inhibitors, indicating that endocytosis plays a major role in this process. Additionally, co-culture experiments with HEK-293 cells expressing red fluorescent protein (RFP)-tagged CD9 and the chondrocytic cell line OUMS-27 confirmed the transfer of RFP-positive EVs across a 600-nm but not a 30-nm filter. These findings suggest that EVs from chondrocytes are released into joint fluid and taken up by cells within the cartilage, potentially facilitating communication between cartilage and synovium. The results underscore the importance of EVs in OA pathophysiology.
Collapse
Affiliation(s)
- Takashi Ohtsuki
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.O.); (I.S.); (K.I.); (G.O.); (S.W.)
| | - Ikumi Sato
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.O.); (I.S.); (K.I.); (G.O.); (S.W.)
| | - Ren Takashita
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.O.); (I.S.); (K.I.); (G.O.); (S.W.)
| | - Shintaro Kodama
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.O.); (I.S.); (K.I.); (G.O.); (S.W.)
| | - Kentaro Ikemura
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.O.); (I.S.); (K.I.); (G.O.); (S.W.)
| | - Gabriel Opoku
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.O.); (I.S.); (K.I.); (G.O.); (S.W.)
| | - Shogo Watanabe
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.O.); (I.S.); (K.I.); (G.O.); (S.W.)
| | - Takayuki Furumatsu
- Department of Orthopedic Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.F.); (K.N.)
| | - Hiroshi Yamada
- Department of Neuroscience, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan;
| | - Mitsuru Ando
- Laboratory of Biomaterials, Institute for Life and Medical Sciences, Kyoto University, Kawahara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan;
| | - Kazunari Akiyoshi
- Department of Immunology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan;
| | - Keiichiro Nishida
- Department of Orthopedic Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.F.); (K.N.)
| | - Satoshi Hirohata
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (T.O.); (I.S.); (K.I.); (G.O.); (S.W.)
| |
Collapse
|
6
|
Voos JE, Moyal A, Furdock R, Caplan AI, Bonfield TL, Calcei JG. Culture Expansion Alters Human Bone Marrow-Derived Mesenchymal Stem Cell Production of Osteoarthritis-Relevant Cytokines and Growth Factors. Arthroscopy 2024:S0749-8063(24)00875-2. [PMID: 39505158 DOI: 10.1016/j.arthro.2024.10.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024]
Abstract
PURPOSE The purposes of this study were to characterize the human bone marrow-derived mesenchymal stem cells (BM-MSCs) production of osteoarthritis-relevant cytokines and growth factors as they are purified and multiplied, a process termed culture expansion, and to compare the immunomodulatory potential of BM-MSCs based on source and medium used for culture expansion. METHODS BM-MSCs were obtained from iliac crest bone marrow aspirates of 4 healthy donors. These 4 BM-MSC cell lines underwent 4 rounds, or "passages," of the institutional culture expansion protocol, using institutional culture media. The secretory molecules known to play a role in osteoarthritis-related inflammatory immune response, cartilage degradation, and patient symptoms, together called the BM-MSC "secretome," were measured at each passage. Three lines of commercially available BM-MSCs from healthy donors underwent culture expansion by the same protocol, using commercial culture media. The commercial BM-MSCs secretome and the institutional BM-MSCs secretome were compared at each passage. Significance was set at P < .05. RESULTS Institutional BM-MSCs produced less interleukin-6 at passages 3 (237 ± 113 pg/mL) and 4 (237 ± 113 pg/mL) compared with passages 1 (884 ± 97 pg/mL) and 2 (1071 ± 129 pg/mL; P < .01). Institutional BM-MSCs produced more macrophage inflammatory protein 3-alpha at passage 4 than at passage 1 (106 ± 41 vs 32 ± 7 pg/mL; P < .01). Across passages of culture expansion, institutional BM-MSCs grown on institutional medium expressed more interleukin-6 (P < .001), interleukin-10 (P < .001), interleukin-1 beta (P < .001), tumor necrosis factor alpha (P = .004), and vascular endothelial growth factor C (P = .003) than commercially available BM-MSCs grown on commercial medium. CONCLUSIONS Culture expansion alters key molecules within the BM-MSC secretome. Additionally, differences in BM-MSC source and culture medium alter the BM-MSC secretome and its immunomodulatory potential. CLINICAL RELEVANCE This study characterizes the in-vitro changes in BM-MSC secretome during culture expansion based on the cell source and culture medium. It suggests nonequivalence of culture-expanded BM-MSC therapies obtained from different donors using different culture media, even if delivering equivalent numbers of BM-MSCs.
Collapse
Affiliation(s)
- James E Voos
- University Hospitals Drusinsky Sports Medicine Institute, Cleveland, Ohio, U.S.A.; Case Western Reserve University School of Medicine (CWRU SOM), CWRU College of Arts and Sciences, Cleveland, Ohio, U.S.A
| | - Andrew Moyal
- University Hospitals Drusinsky Sports Medicine Institute, Cleveland, Ohio, U.S.A.; Case Western Reserve University School of Medicine (CWRU SOM), CWRU College of Arts and Sciences, Cleveland, Ohio, U.S.A..
| | - Ryan Furdock
- University Hospitals Drusinsky Sports Medicine Institute, Cleveland, Ohio, U.S.A.; Case Western Reserve University School of Medicine (CWRU SOM), CWRU College of Arts and Sciences, Cleveland, Ohio, U.S.A
| | - Arnold I Caplan
- Case Western Reserve University School of Medicine (CWRU SOM), CWRU College of Arts and Sciences, Cleveland, Ohio, U.S.A.; Department of Biology, Case Western Reserve University, Cleveland, Ohio, U.S.A.; Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio, U.S.A.; National Center of Regenerative Medicine, Cleveland, Ohio, U.S.A
| | - Tracey L Bonfield
- Case Western Reserve University School of Medicine (CWRU SOM), CWRU College of Arts and Sciences, Cleveland, Ohio, U.S.A.; Department of Biology, Case Western Reserve University, Cleveland, Ohio, U.S.A.; Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio, U.S.A.; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, U.S.A.; National Center of Regenerative Medicine, Cleveland, Ohio, U.S.A
| | - Jacob G Calcei
- University Hospitals Drusinsky Sports Medicine Institute, Cleveland, Ohio, U.S.A.; Case Western Reserve University School of Medicine (CWRU SOM), CWRU College of Arts and Sciences, Cleveland, Ohio, U.S.A
| |
Collapse
|
7
|
Yang H, Zhou Y, Ying B, Dong X, Qian Q, Gao S. Effects of human umbilical cord mesenchymal stem cell-derived exosomes in the rat osteoarthritis models. Stem Cells Transl Med 2024; 13:803-811. [PMID: 38913985 PMCID: PMC11328936 DOI: 10.1093/stcltm/szae031] [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: 08/22/2023] [Accepted: 02/16/2024] [Indexed: 06/26/2024] Open
Abstract
Mesenchymal stem cells (MSCs) offer great potential for treatment of osteoarthritis (OA) by promoting articular cartilage regeneration via paracrine secretion of exosomes; however, the underlying mechanisms are not fully understood. This study aimed to explore the therapeutic effects of exosomes secreted by human umbilical cord-derived MSCs (hUC-MSCs) in rat models of OA and reveal the underlying mechanisms. UC-MSCs and UC-MSC-exosomes were prepared and identified by transmission electron microscopy and flow cytometry. IL-1β-induced OA chondrocytes and the operation and collagenase-induced OA rat models were established. The results of micro-computed tomography, histology, and immunohistochemistry showed that UC-MSC-exosomes promoted cartilage regeneration in OA rats. ELISA results showed that the levels of synovial fluid cytokines, TNF-α, IL-1β, and IL-6, were lower in exosome therapy group than control group in both OA rat models. Exosome treatment significantly downregulated the expression of MMP-13 and ADAMTS-5 in chondrocytes stimulated by IL-1β, and upregulated collagen II expression. These findings suggest that hUC-MSC-exosomes offer a promising option for the therapy for OA.
Collapse
Affiliation(s)
- Huanfeng Yang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, People's Republic of China
- Department of R&D, Oricell Therapeutics, Shanghai, 201203, People's Republic of China
| | - Yiqin Zhou
- Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - Bi Ying
- Department of R&D, Oricell Therapeutics, Shanghai, 201203, People's Republic of China
| | - Xuhui Dong
- Department of R&D, Oricell Therapeutics, Shanghai, 201203, People's Republic of China
| | - Qirong Qian
- Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - Shaorong Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, People's Republic of China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translation Research Center, Shanghai First Maternity and Infant Hospital, School of Life Science and Technology, Tongji University, Shanghai, 201204, People's Republic of China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, People's Republic of China
| |
Collapse
|
8
|
Ibrahiem B, Shamma R, Salama A, Refai H. Magnetic targeting of lornoxicam/SPION bilosomes loaded in a thermosensitive in situ hydrogel system for the management of osteoarthritis: Optimization, in vitro, ex vivo, and in vivo studies in rat model via modulation of RANKL/OPG. Drug Deliv Transl Res 2024; 14:1982-2002. [PMID: 38158473 PMCID: PMC11153292 DOI: 10.1007/s13346-023-01503-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
Osteoarthritis is a bone and joint condition characterized pathologically by articular cartilage degenerative damage and can develop into a devastating and permanently disabling disorder. This investigation aimed to formulate the anti-inflammatory drug lornoxicam (LOR) into bile salt-enriched vesicles loaded in an in situ forming hydrogel as a potential local treatment of osteoarthritis. This was achieved by formulating LOR-loaded bilosomes that are also loaded with superparamagnetic iron oxide nanoparticles (SPIONs) for intra-muscular (IM) administration to improve joint targeting and localization by applying an external magnet to the joint. A 31.22 full factorial design was employed to develop the bilosomal dispersions and the optimized formula including SPION (LSB) was loaded into a thermosensitive hydrogel. Moreover, in vivo evaluation revealed that the IM administration of LSB combined with the application of an external magnet to the joint reversed carrageen-induced suppression in motor activity and osteoprotegerin by significantly reducing the elevations in mitogen-activated protein kinases, extracellular signal-regulated kinase, and receptor activator of nuclear factor kappa beta/osteoprotegerin expressions. In addition, the histopathological evaluation of knee joint tissues showed a remarkable improvement in the injured joint tissues. The results proved that the developed LSB could be a promising IM drug delivery system for osteoarthritis management.
Collapse
Affiliation(s)
- Basma Ibrahiem
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, 12566, Egypt
| | - Rehab Shamma
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, El-Kasr El-Aini Street, Cairo, 11562, Egypt
| | - Abeer Salama
- Department of Pharmacology, National Research Centre (NRC), Giza, 12622, Egypt
| | - Hanan Refai
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, 12566, Egypt.
| |
Collapse
|
9
|
Shin JS, Lee H, Kim SH, Noh KC, Kim SJ, Kim HN, Choi JY, Song SY. Identification of plasma and urinary inflammatory markers in severe knee osteoarthritis: Relations with synovial fluid markers. Knee Surg Relat Res 2024; 36:19. [PMID: 38773579 PMCID: PMC11106897 DOI: 10.1186/s43019-024-00223-8] [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: 01/23/2024] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND This study aimed to identify plasma and urinary cytokines as potential biomarkers for severe knee osteoarthritis (OA). It also investigated associations between these cytokines and cartilage markers, as well as their connections with synovial fluid (SF) markers. METHODS Samples of plasma, urine, and SF were obtained from patients (n = 40) undergoing total knee arthroplasty (TKA) or unicompartmental knee arthroplasty (UKA) due to severe knee OA. Control samples of plasma and urine were collected from non-OA individuals (n = 15). We used a Luminex immunoassay for the simultaneous measurement of 19 cytokines, MMP-1, and MMP-3 levels. COMP, CTX-II, and hyaluronan (HA) levels were quantified using enzyme-linked immunosorbent assay (ELISA) kits. Receiver operating characteristic (ROC) curves were utilized to analyze each biomarker's performance. Correlations among these biomarkers were evaluated via Spearman's correlation. RESULTS The levels of plasma (p)CCL11, pCXCL16, pIL-8, pIL-15, pHA, urinary (u)CCL2, uCCL11, uCCL19, uCXCL16, uIL-1β, uIL-6, uIL-8, uIL-12p70, uIL-15, uIL-33, uMMP-3, uHA, uCTX-II, and uCOMP were significantly elevated in individuals with severe knee OA. Notably, specific correlations were observed between the plasma/urine biomarkers and SF biomarkers: pCCL11 with sfHA (r = 0.56) and sfTNF-α (r = 0.58), pIL-15 with sfCCL19 (r = 0.43) and sfCCL20 (r = 0.44), and uCCL19 with sfCCL11 (r = 0.45) and sfIL-33 (r = 0.51). Positive correlations were also observed between uCCL11 and its corresponding sfCCL11(r = 0.49), as well as between sfCCL11 and other cytokines, namely sfCCL4, sfCCL19, sfCCL20, sfIL-33, and sfTNF-α (r = 0.46-0.63). CONCLUSION This study provides an extensive profile of systemic inflammatory mediators in plasma of knee OA and identified four inflammatory markers (pCCL11, pIL-15, uCCL11, and uCCL19) reflecting joint inflammation.
Collapse
Affiliation(s)
- Ji-Sun Shin
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, 7, Keunjaebong-gil, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea
| | - Hyobeom Lee
- Department of Orthopaedic Surgery, Kangdong Sacred Heart Hospital, 150, Seongan-ro, Gangdong-gu, Seoul, 05355, Republic of Korea
| | - Seong Hyeon Kim
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, 7, Keunjaebong-gil, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea
| | - Kyu-Cheol Noh
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, 7, Keunjaebong-gil, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea
| | - Sung Jae Kim
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, 7, Keunjaebong-gil, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea
| | - Hyong Nyun Kim
- Department of Orthopedic Surgery, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, 1, Singil-ro, Yeongdeungpo-gu, Seoul, 07441, Republic of Korea
| | - Jae-Young Choi
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Si Young Song
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, 7, Keunjaebong-gil, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea.
| |
Collapse
|
10
|
Sapoval M, Querub C, Pereira H, Pellerin O, Boeken T, Di Gaeta A, Ahmar MA, Lefevre-Colau MM, Nguyen C, Daste C, Lacroix M, Laredo JD, Sabatier B, Martelli N, Chatellier G, Déan C, Rannou F. Genicular artery embolization for knee osteoarthritis: Results of the LipioJoint-1 trial. Diagn Interv Imaging 2024; 105:144-150. [PMID: 38102013 DOI: 10.1016/j.diii.2023.12.003] [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: 09/27/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
PURPOSE The purpose of this study was to evaluate the safety and efficacy of transient genicular artery embolization (GAE) using an ethiodized oil-based emulsion for the treatment of knee osteoarthritis (KOA). MATERIALS AND METHODS This prospective, single-arm, open-label, multicenter, first-in-human cohort trial was registered on ClinicalTrials.gov (NCT04733092). The main inclusion criterion was diagnosis of KOA according to a visual analogue scale (VAS) pain score ≥ 40 mm (score range: 0-100 mm), despite conservative treatment for at least three months. Treatment efficacy was assessed using changes in VAS pain score, Mean Western Ontario & McMaster Universities osteoarthritis (WOMAC) function score (normalized to 100; score ranging from 0 to100) and outcome measures in rheumatoid arthritis clinical trials (OMERACT)-Osteoarthritis Research Society (OARSI) set of responder criteria. RESULTS Twenty-two consecutive participants (13 women; mean age, 66 ± 9 [standard deviation (SD)]) were included and underwent GAE. Emulsion consisted in a mixture of ioversol and ethiodized oil (ratio 1:3, respectively) prepared extemporaneously. The rate of serious adverse events attributed to GAE within one month was 5% (1/22), corresponding to reversible worsening of renal function. Immediate technical success rate was 100%. Mean VAS pain score dropped from 74.4 ± 16.5 (SD) mm at baseline to 37.2 ± 26.7 (SD) mm at three months (P < 0.001). Mean WOMAC function score (normalized to 100: score ranging from 0 to 100) decreased from 57.3 ± 17.1 (SD) at baseline to 33.5 ± 25.9 (SD) at three months (P < 0.001). At three months, 16 out of 22 participants (73%) were considered responders according to the OMERACT-OARSI set of responder criteria, including high improvement in either pain or WOMAC function, or improvement in both pain and WOMAC function. CONCLUSION GAE using an ethiodized oil-based emulsion is safe and improves pain and function in participants with KOA for at least three months.
Collapse
Affiliation(s)
- Marc Sapoval
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Vascular and Oncological Interventional Radiology, 75015 Paris, France; Université Paris Cité, PARCC - INSERM Unité-970, 75015 Paris, France.
| | - Charles Querub
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Vascular and Oncological Interventional Radiology, 75015 Paris, France
| | - Helena Pereira
- INSERM, Centre d'investigation Clinique 1418 Épidémiologie Clinique, 75015 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, Clinical Research Unit, 75015 Paris, France
| | - Olivier Pellerin
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Vascular and Oncological Interventional Radiology, 75015 Paris, France; Université Paris Cité, PARCC - INSERM Unité-970, 75015 Paris, France
| | - Tom Boeken
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Vascular and Oncological Interventional Radiology, 75015 Paris, France; HeKA team, INRIA, 75012 Paris, France
| | - Alessandro Di Gaeta
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Vascular and Oncological Interventional Radiology, 75015 Paris, France
| | - Marc Al Ahmar
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Vascular and Oncological Interventional Radiology, 75015 Paris, France
| | - Marie-Martine Lefevre-Colau
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Service de Rééducation et de Réadaptation de l'Appareil Locomoteur et des Pathologies du Rachis, 75014 Paris, France; Université de Paris Cité, INSERM UMR-S 1153, Centre de Recherche Épidémiologie Et Statistique Paris (CRESS), ECaMO Team, 75004 Paris, France
| | - Christelle Nguyen
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Service de Rééducation et de Réadaptation de l'Appareil Locomoteur et des Pathologies du Rachis, 75014 Paris, France; Université Paris Cité, INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs (T3S), Campus Saint-Germain-des-Prés, 75006 Paris, France
| | - Camille Daste
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Service de Rééducation et de Réadaptation de l'Appareil Locomoteur et des Pathologies du Rachis, 75014 Paris, France; Université de Paris Cité, INSERM UMR-S 1153, Centre de Recherche Épidémiologie Et Statistique Paris (CRESS), ECaMO Team, 75004 Paris, France
| | - Maxime Lacroix
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, Service d'imagerie, 75015 Paris, France
| | - Jean-Denis Laredo
- Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Service de Chirurgie Orthopédique Et Traumatologique, 75010 Paris, France
| | - Brigitte Sabatier
- HeKA team, INRIA, 75012 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, Pharmacie, 75015 Paris, France; Université Paris-Saclay, Faculté de Pharmacie, Pharmacie Clinique, 91400 Orsay, France
| | - Nicolas Martelli
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, Pharmacie, 75015 Paris, France; Université Paris-Saclay, Faculté de Pharmacie, GRADES, 91400 Orsay, France
| | - Gilles Chatellier
- INSERM, Centre d'investigation Clinique 1418 Épidémiologie Clinique, 75015 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, Clinical Research Unit, 75015 Paris, France
| | - Carole Déan
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Vascular and Oncological Interventional Radiology, 75015 Paris, France
| | - François Rannou
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Service de Rééducation et de Réadaptation de l'Appareil Locomoteur et des Pathologies du Rachis, 75014 Paris, France; Université Paris Cité, INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs (T3S), Campus Saint-Germain-des-Prés, 75006 Paris, France
| |
Collapse
|
11
|
Zhou R, Zhao L, Wang Q, Cheng Y, Song M, Huang C. Plasma microRNA-320c as a Potential Biomarker for the Severity of Knee Osteoarthritis and Regulates cAMP Responsive Element Binding Protein 5 (CREB5) in Chondrocytes. DISEASE MARKERS 2024; 2024:9936295. [PMID: 38549717 PMCID: PMC10973101 DOI: 10.1155/2024/9936295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/08/2024] [Accepted: 03/08/2024] [Indexed: 04/02/2024]
Abstract
Objective Osteoarthritis (OA) is a commonly known prevalent joint disease, with limited therapeutic methods. This study aimed to investigate the expression of plasma microRNA-320c (miR-320c) in patients with knee OA and to explore the clinical value and potential mechanism of miR-320c in knee OA. Methods Forty knee OA patients and 20 healthy controls were enrolled. The levels of plasma miR-320c and plasma inflammatory cytokines were measured by real-time PCR or ELISA. Correlations of Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores and cytokine levels with the miR-320c expression level were evaluated by Pearson correlation analysis. Then, a receiver operating characteristic (ROC) curve was drawn to analyse the diagnostic value of miR-320c in OA. Finally, the interaction of miR-320c and cAMP responsive element binding protein 5 (CREB5) was determined using a luciferase reporter assay, and the effect of CREB5 on the cAMP pathway was assessed. Results The expression level of plasma miR-320c was significantly higher in OA patients than in healthy controls (p < 0.05). The increased plasma miR-320c level was positively correlated with the WOMAC score (r = 0.796, p < 0.001) and the plasma interleukin (IL)-1β (r = 0.814, p < 0.001) and IL-6 (r = 0.695, p < 0.001) levels in patients with OA. ROC curve analysis demonstrated the relatively high diagnostic accuracy of plasma miR-320c for OA. Furthermore, the luciferase reporter assay results showed that miR-320c regulates CREB5 expression by binding to the CREB5 3'-untranslated region. Moreover, suppression of CREB5 significantly reduced the expression levels of c-fos and c-jun. Conclusion Our results indicate that plasma miR-320c may serve as a potential novel predictor of the severity of knee OA and that miR-320c may play an important role in the pathogenesis of OA through inhibiting the cAMP pathway by targeting CREB5.
Collapse
Affiliation(s)
- Rongwei Zhou
- Department of Respiratory and Critical Care Medicine, School of Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, China
| | - Like Zhao
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qian Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yongjing Cheng
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Miao Song
- Department of Emergency Medicine, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Cibo Huang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
12
|
Giordo R, Tulasigeri Totiger S, Caggiari G, Cossu A, Manunta AF, Posadino AM, Pintus G. Protective Effect of Knee Postoperative Fluid on Oxidative-Induced Damage in Human Knee Articular Chondrocytes. Antioxidants (Basel) 2024; 13:188. [PMID: 38397786 PMCID: PMC10886415 DOI: 10.3390/antiox13020188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The oxidative-stress-elicited deterioration of chondrocyte function is the initial stage of changes leading to the disruption of cartilage homeostasis. These changes entail a series of catabolic damages mediated by proinflammatory cytokines, MMPs, and aggrecanases, which increase ROS generation. Such uncontrolled ROS production, inadequately balanced by the cellular antioxidant capacity, eventually contributes to the development and progression of chondropathies. Several pieces of evidence show that different growth factors, single or combined, as well as anti-inflammatory cytokines and chemokines, can stimulate chondrogenesis and improve cartilage repair and regeneration. In this view, hypothesizing a potential growth-factor-associated action, we investigate the possible protective effect of post-operation knee fluid from patients undergoing prosthesis replacement surgery against ROS-induced damage on normal human knee articular chondrocytes (HKACs). To this end, HKACs were pre-treated with post-operation knee fluid and then exposed to H2O2 to mimic oxidative stress. Intracellular ROS levels were measured by using the molecular probe H2DCFDA; cytosolic and mitochondrial oxidative status were assessed by using HKACs infected with lentiviral particles harboring the redox-sensing green fluorescent protein (roGFP); and cell proliferation was determined by measuring the rate of DNA synthesis with BrdU incorporation. Moreover, superoxide dismutase (SOD), catalase, and glutathione levels from the cell lysates of treated cells were also measured. Postoperative peripheral blood sera from the same patients were used as controls. Our study shows that post-operation knee fluid can counteract H2O2-elicited oxidative stress by decreasing the intracellular ROS levels, preserving the cytosolic and mitochondrial redox status, maintaining the proliferation of oxidatively stressed HKACs, and upregulating chondrocyte antioxidant defense. Overall, our results support and propose an important effect of post-operation knee fluid substances in maintaining HKAC function by mediating cell antioxidative system upregulation and protecting cells from oxidative stress.
Collapse
Affiliation(s)
- Roberta Giordo
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
| | - Smitha Tulasigeri Totiger
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
| | - Gianfilippo Caggiari
- Orthopaedic and Traumatology Department, University Hospital, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.C.); (A.F.M.)
| | - Annalisa Cossu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
| | - Andrea Fabio Manunta
- Orthopaedic and Traumatology Department, University Hospital, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.C.); (A.F.M.)
| | - Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
- Department of Medical Laboratory Sciences, College of Health Sciences, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
13
|
Hengtrakool P, Leearamwat N, Sengprasert P, Wongphoom J, Chaichana T, Taweevisit M, Ngarmukos S, Tanavalee A, Palaga T, Reantragoon R. Infrapatellar fat pad adipose tissue-derived macrophages display a predominant CD11c+CD206+ phenotype and express genotypes attributable to key features of OA pathogenesis. Front Immunol 2024; 15:1326953. [PMID: 38361943 PMCID: PMC10867170 DOI: 10.3389/fimmu.2024.1326953] [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: 10/24/2023] [Accepted: 01/03/2024] [Indexed: 02/17/2024] Open
Abstract
Objectives In knee osteoarthritis (OA), macrophages are the most predominant immune cells that infiltrate synovial tissues and infrapatellar fat pads (IPFPs). Both M1 and M2 macrophages have been described, but their role in OA has not been fully investigated. Therefore, we investigated macrophage subpopulations in IPFPs and synovial tissues of knee OA patients and their correlation with disease severity, examined their transcriptomics, and tested for factors that influenced their polarization. Methods Synovial tissues and IPFPs were obtained from knee OA patients undergoing total knee arthroplasty. Macrophages isolated from these joint tissues were characterized via flow cytometry. Transcriptomic profiling of each macrophage subpopulations was performed using NanoString technology. Peripheral blood monocyte-derived macrophages (MDMs) were treated with synovial fluid and synovial tissue- and IPFP-conditioned media. Synovial fluid-treated MDMs were treated with platelet-rich plasma (PRP) and its effects on macrophage polarization were observed. Results Our findings show that CD11c+CD206+ macrophages were predominant in IPFPs and synovial tissues compared to other macrophage subpopulations (CD11c+CD206-, CD11c-CD206+, and CD11c-CD206- macrophages) of knee OA patients. The abundance of macrophages in IPFPs reflected those in synovial tissues but did not correlate with disease severity as determined from Mankin scoring of cartilage destruction. Our transcriptomics data demonstrated highly expressed genes that were related to OA pathogenesis in CD11c+CD206+ macrophages than CD11c+CD206-, CD11c-CD206+, and CD11c-CD206- macrophages. In addition, MDMs treated with synovial fluid, synovial tissue-conditioned media, or IPFP-conditioned media resulted in different polarization profiles of MDMs. IPFP-conditioned media induced increases in CD86+CD206+ MDMs, whereas synovial tissue-conditioned media induced increases in CD86+CD206- MDMs. Synovial fluid treatment (at 1:8 dilution) induced a very subtle polarization in each macrophage subpopulation. PRP was able to shift macrophage subpopulations and partially reverse the profiles of synovial fluid-treated MDMs. Conclusion Our study provides an insight on the phenotypes and genotypes of macrophages found in IPFPs and synovial tissues of knee OA patients. We also show that the microenvironment plays a role in driving macrophages to polarize differently and shifting macrophage profiles can be reversed by PRP.
Collapse
Affiliation(s)
- Patchanika Hengtrakool
- Medical Microbiology Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Nitigorn Leearamwat
- Medical Microbiology Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Panjana Sengprasert
- Immunology Division, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jutamas Wongphoom
- Department of Pathology, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Thiamjit Chaichana
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Mana Taweevisit
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Srihatach Ngarmukos
- Department of Orthopedics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Biologics for Knee Osteoarthritis Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Aree Tanavalee
- Department of Orthopedics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Biologics for Knee Osteoarthritis Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Rangsima Reantragoon
- Immunology Division, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Skeletal Disorders and Enzyme Reaction Mechanism, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
14
|
Meng J, Cai Y, Yao J, Yan H. Bidirectional causal relationship between psychiatric disorders and osteoarthritis: A univariate and multivariate Mendelian randomization study. Brain Behav 2024; 14:e3429. [PMID: 38361326 PMCID: PMC10869882 DOI: 10.1002/brb3.3429] [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: 08/04/2023] [Revised: 11/11/2023] [Accepted: 01/27/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Observational studies have shown associations between psychiatric disorders and osteoarthritis (OA). However, the causal impact of different psychiatric disorder types on specific sites of osteoarthritis remains unclear. This study aimed to comprehensively understand the potential causal associations between psychiatric disorders and osteoarthritis using Mendelian randomization (MR) analysis. METHODS We collected data from genome-wide association studies of knee osteoarthritis (KOA) (n = 403,124), hip osteoarthritis (HOA) (n = 393,873), osteoarthritis of the knee or hip (KHOA) (n = 417,596), as well as three psychiatric disorders: bipolar disorder (n = 41,917), major depressive disorder (n = 170,756), and schizophrenia (n = 76,755) among European populations. We applied bidirectional univariate and multivariate MR analyses, including inverse variance weighted, Mendelian randomization-Egger, weighted median, simple mode, and weighted mode. We considered p < .05 as a criterion for identifying potential evidence of association. Bonferroni correction was used for multiple tests. RESULTS Our univariate MR analysis results demonstrated that bipolar disorder is a protective factor for KOA (OR = 0.90, 95% CI = 0.83 to 0.97, p = 0.0048) and may also be protective for KHOA (p = 0.02). Conversely, major depression has a positive causal effect on both KOA (OR = 1.27; 95% CI = 1.08 to 1.49; p = 0.0036) and KHOA (OR = 1.24; 95% CI = 1.12 to 1.37; p = 3.62×10-05 ). Furthermore, our analysis suggested that KHOA may be a risk factor for major depression (OR = 1.06; 95% CI = 1.00 to 1.12; p = 0.0469) in reverse MR. After adjusting smoking (OR = 1.46; 95% CI = 1.19 to 1.65; p = 0.0032) and body mass index (OR = 1.44; 95% CI = 1.09 to 1.81; p = 8.56×10-04 ), the casual association between major depression and KHOA remained. CONCLUSION Our study indicates that major depression is a great risk factor for KHOA, increasing the likelihood of their occurrence. However, further in-depth studies will be required to validate these results and elucidate the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Jinzhi Meng
- Bone and Joint SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Youran Cai
- Department of OphthalmologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jun Yao
- Bone and Joint SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Haiwei Yan
- Department of Sports MedicineThe Fourth Affiliated Hospital of Guangxi Medical UniversityLiuzhouChina
| |
Collapse
|
15
|
Wei L, Pan Q, Teng J, Zhang H, Qin N. Intra-articular administration of PLGA resveratrol sustained-release nanoparticles attenuates the development of rat osteoarthritis. Mater Today Bio 2024; 24:100884. [PMID: 38173866 PMCID: PMC10761803 DOI: 10.1016/j.mtbio.2023.100884] [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: 04/13/2023] [Revised: 11/23/2023] [Accepted: 11/25/2023] [Indexed: 01/05/2024] Open
Abstract
Our previous studies have confirmed that resveratrol (RSV) can prevent the development of osteoarthritis through a variety of mechanisms, such as apoptosis inhibition, autophagy induction and SIRT 1 activation. However, the pharmaceutical application of RSV is mainly limited by its low bioavailability. Here, we designed and synthesized RSV-loaded poly (D, l-lactide-coglycolide acid) (PLGA)-nanoparticles (NPs). The average particle size, polydispersity index and positive charge of RSV-loaded PLGA NPs were 50.40 nm, 0.217 and 12.57 mV, respectively. These nanoparticles had marked encapsulation efficiency (92.35 %) and drug loading (15.1 %) for RSV. It was found that RSV-loaded PLGA NPs not only inhibited the apoptosis of chondrocytes induced by IL-1, but also rescued GAG loss in vitro. Pharmacokinetic data showed that RSV-loaded PLGA NPs demonstrated a significantly profound and prolonged concentration profile in joint tissues, with quantifiable RSV concentrations over 35 days. The therapeutic effects of RSV-loaded PLGA NPs were then examined in rat osteoarthritis models. In vitro magnetic resonance imaging results showed that RSV-loaded PLGA NPs treatment dramatically reduced both T1ρ and T2 relaxation times at 4, 8, 12 weeks during administration, implying that cartilage destruction was alleviated. Histological assessments showed that RSV-loaded PLGA NPs significantly improved osteoarthritis symptoms. Gene expression analysis revealed that osteoarthritis mediator genes were downregulated in rats treated with RSV-PLGA NPs. Mechanistic studies indicated that RSV-loaded PLGA NPs inhibit apoptosis and promote autophagy. Collectively, this study demonstrates that intra-articular delivery of RSV via PLGA NPs might be an effective therapeutic approach for osteoarthritis.
Collapse
Affiliation(s)
- Liwei Wei
- Department of Sports Medicine, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang, Henan, China
| | - Qingqing Pan
- The Third Affiliated Hospital of Xinxiang Medical University, Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Junyan Teng
- Bone Pharmacology Laboratory, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang, Henan, China
| | - Hong Zhang
- Bone Pharmacology Laboratory, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang, Henan, China
| | - Na Qin
- Bone Pharmacology Laboratory, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang, Henan, China
| |
Collapse
|
16
|
Brognara L, Luna OC, Traina F, Cauli O. Inflammatory Biomarkers and Gait Impairment in Older Adults: A Systematic Review. Int J Mol Sci 2024; 25:1368. [PMID: 38338653 PMCID: PMC10855721 DOI: 10.3390/ijms25031368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/15/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Peripheral inflammation and gait speed alterations are common in several neurological disorders and in the aging process, but the association between the two is not well established. The aim of this systematic literary review is to determine whether proinflammatory markers are a positive predictor for gait impairments and their complications, such as falls in older adults, and may represent a risk factor for slow gait speed and its complications. The systematic review was performed in line with the Preferred Report Items for Systematic Review and Meta-Analyses (PRISMA). A protocol for literature searches was structured a priori and designed according to the International Perspective Register of Systemic Review (PROSPERO: CRD42023451108). Peer-reviewed original articles were identified by searching seven electronic databases: Excerpta Medica Database (EMBASE), SciVerse (ScienceDirect), Scopus, PubMed, Medline, Web of Science, and the Cochrane Library. The search strategy was formulated based on a combination of controlled descriptors and/or keywords related to the topic and a manual search was conducted of the reference lists from the initially selected studies to identify other eligible studies. The studies were thoroughly screened using the following inclusion criteria: older adults, spatiotemporal gait characteristics, and proinflammatory markers. A meta-analysis was not performed due to the heterogeneity of the studies, and the results were narratively synthesized. Due to the clinical and methodological heterogeneity, the studies were combined in a narrative synthesis, grouped by the type of biomarkers evaluated. A standardized data extraction form was used to collect the following methodological outcome variables from each of the included studies: author, year, population, age, sample size, spatiotemporal gait parameters such as gait velocity, and proinflammatory markers such as TNF-α, high sensitivity C-reactive (CRP) proteins, and IL-6. We included 21 out of 51 studies in our review, which examined the association between inflammatory biomarkers and gait impairment. This review highlights the role of TNF-α, CRP, and IL-6 in gait impairment. Biomarkers play an important role in the decision-making process, and IL-6 can be an effective biomarker in establishing the diagnosis of slow gait speed. Further longitudinal research is needed to establish the use of molecular biomarkers in monitoring gait impairment.
Collapse
Affiliation(s)
- Lorenzo Brognara
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40123 Bologna, Italy;
| | - Oscar Caballero Luna
- Department of Nursing, University of Valencia, 46010 Valencia, Spain; (O.C.L.); (O.C.)
| | - Francesco Traina
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40123 Bologna, Italy;
| | - Omar Cauli
- Department of Nursing, University of Valencia, 46010 Valencia, Spain; (O.C.L.); (O.C.)
| |
Collapse
|
17
|
Abou-Raya A, Rizk M, AbdelGhani E, AbdelMegid N. Identification of serum micro-RNAs of early knee osteoarthritis in a cohort of Egyptian patients. ALEXANDRIA JOURNAL OF MEDICINE 2023. [DOI: 10.1080/20905068.2022.2140987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Anna Abou-Raya
- Alexandria University, Faculty of Medicine, Internal Medicine, Alexandria, Egypt
| | - Mohamed Rizk
- Alexandria University, Faculty of Medicine, Clinical Pathology, Alexandria, Egypt
| | - Eman AbdelGhani
- Alexandria University, Faculty of Medicine, Internal Medicine, Alexandria, Egypt
| | - Nermen AbdelMegid
- Alexandria University, Faculty of Medicine, Internal Medicine, Alexandria, Egypt
| |
Collapse
|
18
|
Patnaik R, Riaz S, Sivani BM, Faisal S, Naidoo N, Rizzo M, Banerjee Y. Evaluating the potential of Vitamin D and curcumin to alleviate inflammation and mitigate the progression of osteoarthritis through their effects on human chondrocytes: A proof-of-concept investigation. PLoS One 2023; 18:e0290739. [PMID: 38157375 PMCID: PMC10756552 DOI: 10.1371/journal.pone.0290739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/14/2023] [Indexed: 01/03/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder primarily affecting the elderly, characterized by a prominent inflammatory component. The long-term side effects associated with current therapeutic approaches necessitate the development of safer and more efficacious alternatives. Nutraceuticals, such as Vitamin D and curcumin, present promising therapeutic potentials due to their safety, efficacy, and cost-effectiveness. In this study, we utilized a proinflammatory human chondrocyte model of OA to assess the anti-inflammatory properties of Vitamin D and curcumin, with a particular focus on the Protease-Activated Receptor-2 (PAR-2) mediated inflammatory pathway. Employing a robust siRNA approach, we effectively modulated the expression of PAR-2 to understand its role in the inflammatory process. Our results reveal that both Vitamin D and curcumin attenuate the expression of PAR-2, leading to a reduction in the downstream proinflammatory cytokines, such as Tumor Necrosis Factor-alpha (TNF-α), Interleukin 6 (IL-6), and Interleukin 8 (IL-8), implicated in the OA pathogenesis. Concurrently, these compounds suppressed the expression of Receptor Activator of Nuclear Factor kappa-Β Ligand (RANKL) and its receptor RANK, which are associated with PAR-2 mediated TNF-α stimulation. Additionally, Vitamin D and curcumin downregulated the expression of Interferon gamma (IFN-γ), known to elevate RANKL levels, underscoring their potential therapeutic implications in OA. This study, for the first time, provides evidence of the mitigating effect of Vitamin D and curcumin on PAR-2 mediated inflammation, employing an siRNA approach in OA. Thus, our findings pave the way for future research and the development of novel, safer, and more effective therapeutic strategies for managing OA.
Collapse
Affiliation(s)
- Rajashree Patnaik
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Sumbal Riaz
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Bala Mohan Sivani
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Shemima Faisal
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Nerissa Naidoo
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Yajnavalka Banerjee
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
- Centre for Medical Education, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
19
|
Leite Pereira C, Grad S, Gonçalves RM. Biomarkers for intervertebral disc and associated back pain: From diagnosis to disease prognosis and personalized treatment. JOR Spine 2023; 6:e1280. [PMID: 38156062 PMCID: PMC10751979 DOI: 10.1002/jsp2.1280] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/18/2023] [Accepted: 08/03/2023] [Indexed: 12/30/2023] Open
Abstract
Biomarkers are commonly recognized as objective indicators of a medical state or clinical outcome and have been widely used as clinical and diagnostic tools and surrogate endpoints in many pathological conditions. In the context of intervertebral disc (IVD) and associated back pain, also known as degenerative disc disease (DDD), the use of biomarkers has been poorly explored. DDD is currently diagnosed using imaging techniques and subjective pain scales, limiting an objective association between DDD and pain levels, as well as an evaluation of disease progression. There is a need for objective and reliable measurements for DDD, pain and pathology progression. DDD predictors could also help clinicians in deciding on the optimal treatment for distinct patient groups. This review addresses the current candidate biomarkers in DDD, including imaging, genetic, metabolite and protein-based parameters, both at the tissue and systemic levels, that may become a major advance in the diagnosis and prognosis of the disease, as well as in the management of therapeutic approaches to DDD.
Collapse
Affiliation(s)
- Catarina Leite Pereira
- I3S, Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- INEB, Instituto de Engenharia BiomédicaUniversidade do PortoPortoPortugal
| | | | - Raquel M. Gonçalves
- I3S, Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- INEB, Instituto de Engenharia BiomédicaUniversidade do PortoPortoPortugal
- ICBAS, Instituto de Ciências Biomédicas Abel SalazarUniversidade do PortoPortoPortugal
| |
Collapse
|
20
|
Partan RU, Putra KM, Kusuma NF, Darma S, Reagan M, Muthia P, Radiandina AS, Saleh MGSI, Salim EM. Umbilical Cord Mesenchymal Stem Cell Secretome Improves Clinical Outcomes and Changes Biomarkers in Knee Osteoarthritis. J Clin Med 2023; 12:7138. [PMID: 38002750 PMCID: PMC10672370 DOI: 10.3390/jcm12227138] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
(1) Background: The current treatment for osteoarthritis is ineffective due to its focus on pain relief and lack of cartilage repair. Viscosupplementation such as hyaluronic acid improves symptoms but remains unnoticed for several months. Researchers are exploring cell-based therapies such as mesenchymal stem cells secretome and mesenchymal stem cells, which can repair cartilage damage. The objective of the research is to evaluate and compare the effectiveness of the secretome derived from umbilical cord mesenchymal stem cells (UC-MSCs) with hyaluronic acid (HA). (2) Methods: An open-label clinical trial involving 30 knee osteoarthritis patients divided into two groups received UC-MSC secretome and hyaluronic acid doses. The study assessed clinical outcomes using VAS and WOMAC and measured MMP-3 and TGF-β1 levels before and after treatment. (3) Results: A study of 30 subjects found that the UC-MSC secretome group showed a decrease in pain in the OA knee compared to the HA group. The therapy was most effective after the third injection, and the group showed a decrease in the MMP-3 ratio and an increase in TGF-β1 compared to the hyaluronic acid group. (4) Conclusions: UC-MSC secretome intra-articular injections showed superior clinical improvement, biomarker changes, and no side effects compared to hyaluronic acid over a 5-week interval.
Collapse
Affiliation(s)
- Radiyati Umi Partan
- Internal Medicine Department, Division of Rheumatology, Faculty of Medicine, Sriwijaya University—Dr. Mohammad Hoesin Hospital, Palembang 30126, Indonesia
| | - Khoirun Mukhsinin Putra
- Internal Medicine Department, Division of Rheumatology, Faculty of Medicine, Sriwijaya University—Dr. Mohammad Hoesin Hospital, Palembang 30126, Indonesia
| | - Narisa Felinka Kusuma
- Internal Medicine Department, Division of Rheumatology, Faculty of Medicine, Sriwijaya University—Dr. Mohammad Hoesin Hospital, Palembang 30126, Indonesia
| | - Surya Darma
- Internal Medicine Department, Division of Rheumatology, Faculty of Medicine, Sriwijaya University—Dr. Mohammad Hoesin Hospital, Palembang 30126, Indonesia
| | - Muhammad Reagan
- Internal Medicine Department, Division of Rheumatology, Faculty of Medicine, Sriwijaya University—Dr. Mohammad Hoesin Hospital, Palembang 30126, Indonesia
| | - Putri Muthia
- Internal Medicine Department, Division of Rheumatology, Faculty of Medicine, Sriwijaya University—Dr. Mohammad Hoesin Hospital, Palembang 30126, Indonesia
| | - Afifah Salshabila Radiandina
- Stem Cell and Regenerative Therapies, From Bench to Market Program, Faculty of Life Science & Medicine, King’s College London, London WC2R 2LS, UK
| | - MGS Irsan Saleh
- Department of Pharmacology, Faculty of Medicine, Sriwijaya University, Palembang 30126, Indonesia;
| | - Eddy Mart Salim
- Internal Medicine Department, Division of Allergy & Immunology, Faculty of Medicine, Sriwijaya University—Dr. Mohammad Hoesin Hospital, Palembang 30126, Indonesia
| |
Collapse
|
21
|
Matejova J, Fecskeova LK, Slovinska L, Harvanova D, Spakova T, Bzdilova J. Plasma-derived extracellular vesicle surface markers CD45, CD326 and CD56 correlate with the stage of osteoarthritis: a primary study of a novel and promising diagnostic tool of the disease. Sci Rep 2023; 13:20071. [PMID: 37973964 PMCID: PMC10654566 DOI: 10.1038/s41598-023-47074-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: 07/25/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Recently, there is a growing interest in the research based on extracellular vesicles (EVs) which represent paracrine factors secreted by almost all cell types. Both, normal and pathological cells are able to release various types of EVs with different physiological properties, functions and compositions. EVs play an important role in intercellular communication, mechanism and tissue repair. Moreover, EVs could help not only in the treatment of diseases but also in their diagnostics. This work focused on the evaluation of the potential of EVs being used as biomarkers for the diagnosis of osteoarthritis (OA) based on a comparison of the composition of EVs separated from platelet-poor plasma (PPP) of healthy donors and OA patients at different stages of OA. OA is established as a complex syndrome with extensive impact on multiple tissues within the synovial joint. It is a chronic disease of musculoskeletal system that mainly affects the elderly. Depending on the use of the Kellgren-Lawrence classification system, there are four grades of OA which have a negative impact on patients' quality of life. It is very difficult to detect OA in its early stages, so it is necessary to find a new diagnostic method for its timely detection. PPP samples were prepared from whole blood. PPP-EVs were separated from 3 groups of donors-healthy control, early stage OA, end-stage OA, and their content was compared and correlated. EVs from PPP were separated by size exclusion chromatography and characterized in terms of their size, yield and purity by NTA, western blotting, ELISA and flow cytometry. Detection of surface markers expression in EVs was performed using MACSPlex approach. Inflammatory and growth factors in EVs were analysed using MAGPix technology. Our study confirmed significant differences between EVs surface markers of patients and healthy controls correlating with the age of donor (CD63, CD31 and ROR1) and stage of OA (CD45, CD326 and CD56), respectively. Circulating EVs have been under extensive investigation for their capability to predict OA pathology diagnosis as potential targets for biomarker discovery. Taken together, obtained results indicated that PPP-EVs surface markers could be used as potential biomarkers in the early diagnosis of OA.
Collapse
Affiliation(s)
- Jana Matejova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Livia K Fecskeova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Lucia Slovinska
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Denisa Harvanova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Timea Spakova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Jana Bzdilova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia.
| |
Collapse
|
22
|
Wu Y, Ying J, Zhu X, Xu C, Wu L. Pachymic acid suppresses the inflammatory response of chondrocytes and alleviates the progression of osteoarthritis via regulating the Sirtuin 6/NF-κB signal axis. Int Immunopharmacol 2023; 124:110854. [PMID: 37657246 DOI: 10.1016/j.intimp.2023.110854] |