1
|
Liu N, Jiang F, Feng Z, Mei S, Cui Y, Zheng Y, Yang W, Wang B, Zhang W, Xie J, Zhang N. MgO@SiO 2 nanocapsules: a controlled magnesium ion release system for targeted inhibition of osteoarthritis progression. NANOSCALE ADVANCES 2025; 7:1814-1824. [PMID: 39911730 PMCID: PMC11791780 DOI: 10.1039/d4na00900b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/17/2025] [Indexed: 02/07/2025]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by degenerative changes in articular cartilage and chronic inflammation. Recent studies suggest that intra-articular (i.a.) injection of magnesium salts holds promise as a therapeutic approach for OA. However, the rapid diffusion of magnesium ions limits their efficacy, resulting in a short duration of action. To overcome this limitation, we developed a nanoparticle delivery system using MgO@SiO2 core/shell nanoparticles, designed as a depot for the controlled release of magnesium ions. Electron microscopy confirmed the formation of the core/shell structure with silica shells of varying thickness. Release studies demonstrated that the silica coating effectively slows nanoparticle degradation, extending magnesium release to over 72 hours. In a rabbit OA model, i.a. injection of these nanocapsules significantly mitigated the pathological progression of OA within four weeks without inducing systemic toxicity. Immunohistochemical analysis further revealed that MgO@SiO2 nanocapsules alleviate the inflammatory response in OA cartilage by inhibiting the NF-κB/p65 signaling pathway. In summary, this study confirms the potential of intra-articular magnesium supplementation as a therapeutic option for OA and introduces a novel approach to enhance the delivery and efficacy of magnesium ions in OA treatment, addressing a relatively underexplored area in the field.
Collapse
Affiliation(s)
- Na Liu
- Affiliated Xinhua Hospital of Dalian University Dalian Liaoning 116000 China
| | - Fangchao Jiang
- Department of Chemistry of University of Georgia Athens Georgia 30602 USA
| | - Zhizi Feng
- Department of Chemistry of University of Georgia Athens Georgia 30602 USA
| | - Sen Mei
- Affiliated Xinhua Hospital of Dalian University Dalian Liaoning 116000 China
| | - Yingna Cui
- Department of Chemistry of Dalian University Dalian Liaoning 116000 China
| | - Yu Zheng
- Affiliated Xinhua Hospital of Dalian University Dalian Liaoning 116000 China
| | - Wei Yang
- Department of Chemistry of University of Georgia Athens Georgia 30602 USA
| | - Benjie Wang
- Affiliated Xinhua Hospital of Dalian University Dalian Liaoning 116000 China
| | - Weizhong Zhang
- Department of Chemistry of University of Georgia Athens Georgia 30602 USA
| | - Jin Xie
- Department of Chemistry of University of Georgia Athens Georgia 30602 USA
| | - Nan Zhang
- Affiliated Xinhua Hospital of Dalian University Dalian Liaoning 116000 China
| |
Collapse
|
2
|
Ma R, Zhang C, Liu J, Ren J, Huang H, Wang G, Ding Y, Li X. Associations of magnesium depletion score with the incidence and mortality of osteoarthritis: a nationwide study. Front Immunol 2025; 16:1512293. [PMID: 40092981 PMCID: PMC11907003 DOI: 10.3389/fimmu.2025.1512293] [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: 10/16/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Background Magnesium is an essential immune nutrient for the body, and recent studies have found that it plays an important role in osteoarthritis (OA). Magnesium depletion score(MDS) is a new method for evaluating the magnesium status of the body. Our objective is to explore the association between MDS and the incidence of OA, as well as the relationship between MDS and mortality in patients with OA. Methods Eligible participants were obtained from NHANES from 2005 to 2018. Logistic regression models were employed to evaluate the link between MDS and the incidence of OA. Cox regression models were employed to evaluate the link between MDS and mortality among OA patients. In addition, restricted cubic spline was utilized to explore the correlation between MDS and the incidence of OA, as well as the relationship between MDS and mortality in patients with OA. Subgroup analysis were adopted in order to ensure the credibility of the results in different subgroups, including age, gender, race, education level, BMI, smoking, diabetes and hypertension. Results 19,394 individuals qualified for analysis, including 3,256 OA patients. After excluding missing follow-up data, 630 all-cause deaths and 172 cardiovascular deaths (CVDs) were observed in 3,250 OA patients. The individuals with OA had higher levels of MDS. In the logistic regression model, MDS was positively related to OA (MDS≥3 vs. MDS=0, OR =1.83 (1.46-2.30, P<0.001)). Besides, a positive association was observed between MDS and all-cause mortality [MDS≥3 vs. MDS=0, HR =2.56 (1.49-4.41, P<0.001)] and CVDs [MDS≥3 vs. MDS=0, HR =3.00 (1.13-7.98, P=0.01)] in cox regression models. In addition, a 1-unit rise in MDS was significantly linked to an increased risk of mortality. Restricted cubic spline indicated a positive relationship between MDS and incidence and mortality of OA. Subgroup analysis demonstrated that the results are stable in different subgroups. Conclusions MDS is positively correlated with the incidence and mortality of OA. Optimizing the nutritional status of magnesium may bring benefits to OA patients.
Collapse
Affiliation(s)
- Ruicong Ma
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Cheng Zhang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Jiaqing Liu
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Jinyi Ren
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Huina Huang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Guan Wang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Yanchun Ding
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xia Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
3
|
Heffernan SM, Waldron M, Meldrum K, Evans SJ, Conway GE. Red Algae Alters Expression of Inflammatory Pathways in an Osteoarthritis In Vitro Co-Culture. Pharmaceuticals (Basel) 2025; 18:315. [PMID: 40143094 PMCID: PMC11945273 DOI: 10.3390/ph18030315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Osteoarthritis (OA) is one of the most prevalent chronic conditions and significantly contributes to local and global disease burden. Common pharmaceuticals that are used to treat OA cause significant side effects, thus non-pharmaceutical bioactive alternatives have been developed that can impact OA symptoms without severe side-effects. One such alternative is the Red Algae Lithothamnion species (Litho). However, there is little mechanistic knowledge of its potential to effect OA gene expression, and a human in vitro model using commercially available cell lines to test its effectiveness has yet to be developed. Methods: Human osteoblast (hFOB 1.19. CRL-11372) and chondrocyte (C28/I2) cell lines were co-cultured indirectly using transwells. IL1-β was used to induce an inflammatory state and gene expression profiles following treatment were the primary outcome. Conclusions: Results indicated that the model was physiologically relevant, remained viable over at least seven days, untreated or following induction of an inflammatory state while maintaining hFOB 1.19. and C28/I2 cell phenotypic characteristics. Following treatment, Litho reduced the expression of inflammatory and pain associated genes, most notably IL-1β, IL-6, PTGS2 (COX-2) and C1qTNF2 (CTRP2). Confirmatory analysis with droplet digital PCR (ddPCR) revealed that Il-1β induced a significant reduction in C1qTNF2 at 7 days which was ameliorated with Litho treatment. These data present a novel and replicable co-culture model of inflammatory OA that can be used to investigate bioactive nutraceuticals. For the first time, this model demonstrated a reduction in C1qTNF2 expression that was mitigated by Red Algae Lithothamnion species.
Collapse
Affiliation(s)
- Shane M. Heffernan
- Applied Sports Science Technology and Medicine Research Centre (A-STEM), Faculty of Science and Engineering, Swansea University, Swansea SA1 8EN, UK;
| | - Mark Waldron
- Applied Sports Science Technology and Medicine Research Centre (A-STEM), Faculty of Science and Engineering, Swansea University, Swansea SA1 8EN, UK;
| | - Kirsty Meldrum
- In Vitro Toxicology Group, Faculty of Medicine, Health and Life Sciences, Swansea University, Swansea SA2 8PP, UK; (K.M.); (S.J.E.); (G.E.C.)
| | - Stephen J. Evans
- In Vitro Toxicology Group, Faculty of Medicine, Health and Life Sciences, Swansea University, Swansea SA2 8PP, UK; (K.M.); (S.J.E.); (G.E.C.)
| | - Gillian E. Conway
- In Vitro Toxicology Group, Faculty of Medicine, Health and Life Sciences, Swansea University, Swansea SA2 8PP, UK; (K.M.); (S.J.E.); (G.E.C.)
| |
Collapse
|
4
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [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: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
5
|
Han Y, Zheng D, Ji Y, Feng Y, Chen Z, Chen L, Li H, Jiang X, Shen H, Tao B, Zhuang H, Bu W. Active Magnesium Boride/Alginate Hydrogels Rejuvenate Senescent Cells. ACS NANO 2024; 18:23566-23578. [PMID: 39145584 DOI: 10.1021/acsnano.4c07833] [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: 08/16/2024]
Abstract
The clearance of senescent cells may be detrimental to low cell density diseases, such as intervertebral disc degeneration (IVDD), and rejuvenating these cells presents a formidable obstacle. In this study, we investigate a mild-alkalization strategy employing magnesium boride-alginate (MB-ALG) hydrogels to rejuvenate senescent cells associated with age-related diseases. MB-ALG hydrogels proficiently ensnare senescent cells owing to their surface roughness. The hydrolysis of MB-ALG hydrogels liberates hydroxide ions (OH-), effecting a transition from an acidic microenvironment (pH ∼ 6.2) to a mildly alkaline state (pH ∼ 8.0), thereby fostering senescent cell proliferation via activation of the PI3K/Akt/mTOR pathway. Additionally, H2 aids in ROS clearance, which reduces cellular oxidative stress. And, Mg2+ rejuvenates senescent cells by inhibiting Ca2+ influx and fine-tuning the sirt1-p53 signaling pathways. Both in vitro and in vivo experiments conducted on rat intervertebral discs corroborate the sustained antisenescence and rejuvenation properties of MB-ALG hydrogels, with effects persisting for up to 12 weeks postoperation. These discoveries elucidate the role of mild-alkalization in dictating cellular destiny and provide key insights for addressing age-related diseases.
Collapse
Affiliation(s)
- Yingchao Han
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P. R. China
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai 200433, P. R. China
| | - Dandan Zheng
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P. R. China
| | - Yucheng Ji
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P. R. China
| | - Yubo Feng
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P. R. China
| | - Zhanyi Chen
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P. R. China
| | - Lijie Chen
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai 200433, P. R. China
| | - Huiyan Li
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai 200433, P. R. China
| | - Xingwu Jiang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai 200433, P. R. China
| | - Hongxing Shen
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P. R. China
| | - Bangbao Tao
- Department of Neurosurgery, Xinhua Hospital School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P. R. China
| | - Hongjun Zhuang
- Research Center for Translational Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, P. R. China
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Wenbo Bu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
6
|
Mei S, Jiang F, Liu N, Feng Z, Zheng Y, Yang W, Zhang W, Cui Y, Wang W, Xie J, Zhang N. Sol-gel synthesis of magnesium oxide nanoparticles and their evaluation as a therapeutic agent for the treatment of osteoarthritis. Nanomedicine (Lond) 2024; 19:1867-1878. [PMID: 39109508 PMCID: PMC11457622 DOI: 10.1080/17435889.2024.2382421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/16/2024] [Indexed: 10/05/2024] Open
Abstract
Aim: We synthesized MgO NPs via sol-gel reaction and investigated them as carriers to deliver Mg2+ to the affected joint for osteoarthritis (OA).Materials & methods: The physicochemical properties of samples were characterized by transmission electron microscope (TEM), dynamic light scattering (DLS) and x-ray diffraction (XRD). The release of Mg2+ was monitored by ICP-MS. The potential cytotoxicity was evaluated using MTT assay. The efficacy and biosafety were evaluated in a rabbit OA model.Results: MgO NPs can prolong the Mg2+ release time from 0.5 h to 12 h. No significant cytotoxicity was observed when concentrations below 250 μg/ml. Intra-articular samples could effectively alleviate the degeneration and destruction of the cartilage.Conclusion: this study demonstrates the potential of MgO NPs as a safe and effective treatment of OA. Simultaneously, the size of the particles may play a significant role in influencing the therapeutic outcome.
Collapse
Affiliation(s)
- Sen Mei
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Fangchao Jiang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Na Liu
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Zhizi Feng
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Yu Zheng
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Wei Yang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Weizhong Zhang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Yingna Cui
- Department of Chemistry, Dalian University, Dalian, Liaoning, 116000, China
| | - Weiming Wang
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Nan Zhang
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| |
Collapse
|
7
|
Zheng L, Zhao S, Li Y, Xu J, Yan W, Guo B, Xu J, Jiang L, Zhang Y, Wei H, Jiang Q. Engineered MgO nanoparticles for cartilage-bone synergistic therapy. SCIENCE ADVANCES 2024; 10:eadk6084. [PMID: 38457498 PMCID: PMC10923500 DOI: 10.1126/sciadv.adk6084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
The emerging therapeutic strategies for osteoarthritis (OA) are shifting toward comprehensive approaches that target periarticular tissues, involving both cartilage and subchondral bone. This shift drives the development of single-component therapeutics capable of acting on multiple tissues and cells. Magnesium, an element essential for maintaining skeletal health, shows promise in treating OA. However, the precise effects of magnesium on cartilage and subchondral bone are not yet clear. Here, we investigated the therapeutic effect of Mg2+ on OA, unveiling its protective effects on both cartilage and bone at the cellular and animal levels. The beneficial effect on the cartilage-bone interaction is primarily mediated by the PI3K/AKT pathway. In addition, we developed poly(lactic-co-glycolic acid) (PLGA) microspheres loaded with nano-magnesium oxide modified with stearic acid (SA), MgO&SA@PLGA, for intra-articular injection. These microspheres demonstrated remarkable efficacy in alleviating OA in rat models, highlighting their translational potential in clinical applications.
Collapse
Affiliation(s)
- Liming Zheng
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Sheng Zhao
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Yixuan Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Wenjin Yan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Baosheng Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jianbin Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Lifeng Jiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Yifeng Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Hui Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| |
Collapse
|
8
|
Pinto ACMD, de Melo Nunes R, de Freitas Carvalho WV, Girão VCC, Rocha FAC. Systemic and local antiinflammatory effect of magnesium chloride in experimental arthritis. Adv Rheumatol 2024; 64:6. [PMID: 38178208 DOI: 10.1186/s42358-023-00346-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024] Open
Abstract
OBJECTIVE Despite some knowledge gaps in scientific evidence, MgCl2 is largely used for pain relief in musculoskeletal diseases. Mg salts were shown to provide analgesia postoperatively in orthopedic surgery and low Mg levels were linked to arthritis development and severity. We determined the anti-inflammatory activity of MgCl2 in an acute arthritis model. METHODS Mice received 0.1 mg/25µL Zymosan (Zy) or saline into the knees. Joint pain was evaluated using von Frey test; cell influx, and interleukin (IL)-1 level were assessed in joint lavage at 6 h. Synovia were excised for histopathology and analysis of immunoexpression of nuclear factor kappa B (NFκB) and tumor necrosis factor (TNF)-α. Groups (n = 6/group) received either 90 mg/kg MgCl2/100 µL or saline per os (systemic) or 500 µg/25 µL MgCl2 or saline intra-articularly (i.a.) 30 min prior to Zy. RESULTS MgCl2 given either systemically or locally significantly reduced cell influx (p = 0.0012 and p = 0.0269, respectively), pain (p = 0.0005 and p = 0.0038, respectively), and intra-articular IL-1 level (p = 0.0391), as compared to saline. Systemic MgCl2 significantly decreased NFκB (p < 0.05) immmunoexpression, as compared to saline. CONCLUSION MgCl2 given systemically or locally displayed anti-inflammatory activity in a severe acute arthritis model reducing cell influx, pain, and cytokine release. MgCl2 operates at least partially via inhibiting NFκB activation. This is the first in vivo demonstration that MgCl2 decreases cytokine release in arthritis, prompting reduction of inflammation and pain relief.
Collapse
Affiliation(s)
| | - Rodolfo de Melo Nunes
- Departamento de Medicina Interna da Faculdade de Medicina da Universidade Federal do Ceará, Fortaleza - Ceará, Brazil
| | | | | | - Francisco Airton Castro Rocha
- Departamento de Medicina Interna da Faculdade de Medicina da Universidade Federal do Ceará, Fortaleza - Ceará, Brazil.
- Instituto de Biomedicina - Laboratório de Investigação em Osteoartropatias, Rua Coronel Nunes de Melo, 1315 -1º. Andar Rodolfo Teofilo, Fortaleza, CE, CEP: 60430-270, Brazil.
| |
Collapse
|
9
|
Zhang Y, Chen T, Luo P, Li S, Zhu J, Xue S, Cao P, Zhu Z, Li J, Wang X, Wluka AE, Cicuttini F, Ruan G, Ding C. Associations of Dietary Macroelements with Knee Joint Structures, Symptoms, Quality of Life, and Comorbid Conditions in People with Symptomatic Knee Osteoarthritis. Nutrients 2022; 14:3576. [PMID: 36079832 PMCID: PMC9460692 DOI: 10.3390/nu14173576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/20/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA), the most common joint disease in the elderly, has no cure. Macroelements are vital in human health and their relationships with OA are not clear. Clarifying the relationships between macroelements and OA may assist knee OA management. METHODS This study was a post-hoc analysis using data from a two-year randomized controlled trial among 392 participants with knee OA. Dietary macroelements, including calcium, magnesium, potassium, and phosphorus were computed-based on a semi-quantitative food frequency questionnaire at baseline. Knee joint structures (including cartilage volume, cartilage defect, bone marrow lesions, and effusion-synovitis volume), OA symptoms, quality of life, and OA comorbid conditions (including lower limb muscle strength and depressive symptoms) were assessed at baseline and month 24. Western Ontario and McMaster Universities (WOMAC) Index and depressive symptoms were assessed at baseline and months 3, 6, 12, and 24. Quality of life and lower limb muscle strength were assessed at baseline and months 6, 12, and 24. All analyses were conducted using mixed-effects models. RESULTS Higher dietary magnesium and potassium were associated with fewer OA symptoms, higher quality of life, greater lower limb muscle strength, and fewer depressive symptoms, but not with knee joint structures. Higher dietary calcium and phosphorus was not associated with any of the OA-related outcomes, except that dietary phosphorus was associated with greater lower limb muscle strength. CONCLUSIONS In the longitudinal analyses, higher dietary magnesium and potassium intake are associated with fewer OA symptoms, higher quality of life, and milder comorbid conditions in patients with knee OA, suggesting dietary magnesium and potassium may have beneficial effects on OA and could be used for knee OA management.
Collapse
Affiliation(s)
- Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Tianyu Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
| | - Ping Luo
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Department of Spinal Surgery, Changsha Hospital of Hunan Normal University, Changsha 410000, China
| | - Shengfa Li
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Jianwei Zhu
- Department of Orthopedics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Song Xue
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei 230000, China
| | - Peihua Cao
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Jia Li
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Xiaoshuai Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Anita E. Wluka
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3006, Australia
| | - Flavia Cicuttini
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3006, Australia
| | - Guangfeng Ruan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
- Clinical Research Centre, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3006, Australia
- Clinical Research Centre, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| |
Collapse
|
10
|
Wu G, Ma F, Liu Z, Liu J, Xue Y, Zhang M, Wen C, Tang B, Lin L. Hybrid composites with magnesium-containing glycosaminoglycans as a chondroconducive matrix for osteoarthritic cartilage repair. Int J Biol Macromol 2022; 220:1104-1113. [PMID: 35981680 DOI: 10.1016/j.ijbiomac.2022.08.071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/29/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022]
Abstract
The alteration of the extracellular matrix (ECM) homeostasis plays an important role in the development of osteoarthritis (OA). The pathological changes of OA are mainly manifested in the large reduction of components in ECM, like type II collagen and aggrecan, especially hyaluronic acid and chondroitin sulfate and often accompanied by inflammation. Rebuilding ECM and inhibiting inflammation may reverse OA progression. In this work, we developed new magnesium-containing glycosaminoglycans (Mg-GAGs), to create a positive ECM condition for promoting cartilage regeneration and alleviating OA. In vitro results suggested that the introduction of Mg-GAGs contributed to promoting chondrocyte proliferation and facilitated upregulating chondrogenic genes and suppressed inflammation-related factors. Moreover, Mg-GAGs exhibited positive effects on suppressing synovial inflammation, reducing chondrocyte apoptosis and preserving the subchondral bone in the ACLT-induced OA rabbit model. This study provides new insight into ECM-based therapeutic strategy and opens a new avenue for the development of novel OA treatment.
Collapse
Affiliation(s)
- Guofeng Wu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Orthopedics, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, PR China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Fenbo Ma
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Zhengwei Liu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Jiayi Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Yizhebang Xue
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Mengdi Zhang
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China
| | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China; Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, PR China.
| | - Lijun Lin
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
11
|
Bai R, Miao MZ, Li H, Wang Y, Hou R, He K, Wu X, Jin H, Zeng C, Cui Y, Lei G. Increased Wnt/β-catenin signaling contributes to autophagy inhibition resulting from a dietary magnesium deficiency in injury-induced osteoarthritis. Arthritis Res Ther 2022; 24:165. [PMID: 35804467 PMCID: PMC9264717 DOI: 10.1186/s13075-022-02848-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
Background Dietary magnesium deficiency, which is common in modern diet, has been associated with osteoarthritis (OA) susceptibility. Despite this clinical association, no study has addressed if dietary magnesium deficiency accelerates OA development, especially at molecular level. This study aimed to explore aggravating effects of dietary magnesium deficiency on cartilage damage in an injury-induced murine OA model and to determine the underlying mechanism. Methods Twelve-week-old C57BL/6J mice subject to injury-induced OA modeling were randomized into different diet groups in which the mice were fed a diet with daily recommended magnesium content (500 mg/kg) or diets with low magnesium content (100 or 300 mg/kg). Articular cartilage damage was evaluated using the OARSI score. To determine molecular mechanisms in vitro, mouse chondrocytes were treated with media of low magnesium conditions at 0.1 and 0.4 mM, compared with normal magnesium condition at 0.7 mM as control. Anabolic and catabolic factors, autophagy markers, β-catenin, Wnt ligands, and a magnesium channel transient receptor potential cation channel subfamily member 7 (TRPM7) were analyzed by quantitative real-time PCR and immunoblotting. Autolysosomes were detected by DALGreen staining via fluorescence microscopy and autophagosomes were evaluated by transmission electron microscopy. Autophagy markers, β-catenin, and TRPM7 were assessed in vivo in the mouse cartilage, comparing between dietary magnesium deficiency and normal diet, by immunohistochemistry. Results Dietary magnesium deficiency aggravated injury-induced cartilage damage, indicated by significant higher OARSI scores. Autophagy markers LC3-II and Beclin-1 were decreased both in low magnesium diet-fed mice and low magnesium-treated chondrocytes. The number of autolysosomes and autophagosomes was also reduced under low magnesium conditions. Moreover, magnesium deficiency induced decreased anabolic and increased catabolic effect of chondrocytes which could be restored by autophagy activator rapamycin. In addition, reduced autophagy under low magnesium conditions is mediated by activated Wnt/β-catenin signaling. The expression of TRPM7 also decreased in low magnesium diet-fed mice, indicating that downstream changes could be regulated through this channel. Conclusions Dietary magnesium deficiency contributes to OA development, which is mediated by reduced autophagy through Wnt/β-catenin signaling activation. These findings indicated potential benefits of adequate dietary magnesium for OA patients or those individuals at high risk of OA. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02848-0.
Collapse
Affiliation(s)
- Ruijun Bai
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Michael Z Miao
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Curriculum in Oral and Craniofacial Biomedicine, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hui Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Yiqing Wang
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ruixue Hou
- Department of Population Health Science and Policy, Icahn School of Medical at Mount Sinai, New York, NY, USA
| | - Ke He
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Xuan Wu
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Hongyu Jin
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.,Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China.,Hunan Engineering Research Center of Osteoarthritis, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Cui
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. .,Xiangya International Medical Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China. .,Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China. .,Hunan Engineering Research Center of Osteoarthritis, Changsha, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
12
|
Zeng CY, Wang XF, Hua FZ. HIF-1α in Osteoarthritis: From Pathogenesis to Therapeutic Implications. Front Pharmacol 2022; 13:927126. [PMID: 35865944 PMCID: PMC9294386 DOI: 10.3389/fphar.2022.927126] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis is a common age-related joint degenerative disease. Pain, swelling, brief morning stiffness, and functional limitations are its main characteristics. There are still no well-established strategies to cure osteoarthritis. Therefore, better clarification of mechanisms associated with the onset and progression of osteoarthritis is critical to provide a theoretical basis for the establishment of novel preventive and therapeutic strategies. Chondrocytes exist in a hypoxic environment, and HIF-1α plays a vital role in regulating hypoxic response. HIF-1α responds to cellular oxygenation decreases in tissue regulating survival and growth arrest of chondrocytes. The activation of HIF-1α could regulate autophagy and apoptosis of chondrocytes, decrease inflammatory cytokine synthesis, and regulate the chondrocyte extracellular matrix environment. Moreover, it could maintain the chondrogenic phenotype that regulates glycolysis and the mitochondrial function of osteoarthritis, resulting in a denser collagen matrix that delays cartilage degradation. Thus, HIF-1α is likely to be a crucial therapeutic target for osteoarthritis via regulating chondrocyte inflammation and metabolism. In this review, we summarize the mechanism of hypoxia in the pathogenic mechanisms of osteoarthritis, and focus on a series of therapeutic treatments targeting HIF-1α for osteoarthritis. Further clarification of the regulatory mechanisms of HIF-1α in osteoarthritis may provide more useful clues to developing novel osteoarthritis treatment strategies.
Collapse
Affiliation(s)
- Chu-Yang Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xi-Feng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xi-Feng Wang, ; Fu-Zhou Hua,
| | - Fu-Zhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xi-Feng Wang, ; Fu-Zhou Hua,
| |
Collapse
|
13
|
Disease-Modifying Adjunctive Therapy (DMAT) in Osteoarthritis-The Biological Effects of a Multi-Mineral Complex, LithoLexal ® Joint-A Review. Clin Pract 2021; 11:901-913. [PMID: 34940003 PMCID: PMC8700461 DOI: 10.3390/clinpract11040104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/09/2021] [Accepted: 11/19/2021] [Indexed: 01/04/2023] Open
Abstract
Modern advances in molecular medicine have led to the reframing of osteoarthritis as a metabolically active, inflammatory disorder with local and systemic contributing factors. According to the ‘inflammatory theory’ of osteoarthritis, immune response to an initial damage is the key trigger that leads to progressive joint destruction. Several intertwined pathways are known to induce and govern articular inflammation, cartilage matrix degradation, and subchondral bone changes. Effective treatments capable of halting or delaying the progression of osteoarthritis remain elusive. As a result, supplements such as glucosamine and chondroitin sulphate are commonly used despite the lack of scientific consensus. A novel option for adjunctive therapy of osteoarthritis is LithoLexal® Joint, a marine-derived, mineral-rich extract, that exhibited significant efficacy in clinical trials. LithoLexal® has a lattice microstructure containing a combination of bioactive rare minerals. Mechanistic research suggests that this novel treatment possesses various potential disease-modifying properties, such as suppression of nuclear factor kappa-B, interleukin 1β, tumor necrosis factor α, and cyclooxygenase-2. Accordingly, LithoLexal® Joint can be considered a disease-modifying adjunctive therapy (DMAT). LithoLexal® Joint monotherapy in patients with knee osteoarthritis has significantly improved symptoms and walking ability with higher efficacy than glucosamine. Preliminary evidence also suggests that LithoLexal® Joint may allow clinicians to reduce the dose of nonsteroidal anti-inflammatory drugs in osteoarthritic patients by up to 50%. In conclusion, the multi-mineral complex, LithoLexal® Joint, appears to be a promising candidate for DMAT of osteoarthritis, which may narrow the existing gap in clinical practice.
Collapse
|
14
|
Research status of biodegradable metals designed for oral and maxillofacial applications: A review. Bioact Mater 2021; 6:4186-4208. [PMID: 33997502 PMCID: PMC8099919 DOI: 10.1016/j.bioactmat.2021.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/10/2021] [Accepted: 01/10/2021] [Indexed: 01/08/2023] Open
Abstract
The oral and maxillofacial regions have complex anatomical structures and different tissue types, which have vital health and aesthetic functions. Biodegradable metals (BMs) is a promising bioactive materials to treat oral and maxillofacial diseases. This review summarizes the research status and future research directions of BMs for oral and maxillofacial applications. Mg-based BMs and Zn-based BMs for bone fracture fixation systems, and guided bone regeneration (GBR) membranes, are discussed in detail. Zn-based BMs with a moderate degradation rate and superior mechanical properties for GBR membranes show great potential for clinical translation. Fe-based BMs have a relatively low degradation rate and insoluble degradation products, which greatly limit their application and clinical translation. Furthermore, we proposed potential future research directions for BMs in the oral and maxillofacial regions, including 3D printed BM bone scaffolds, surface modification for BMs GBR membranes, and BMs containing hydrogels for cartilage regeneration, soft tissue regeneration, and nerve regeneration. Taken together, the progress made in the development of BMs in oral and maxillofacial regions has laid a foundation for further clinical translation.
Collapse
|
15
|
Dąbrowski M, Zioła-Frankowska A, Frankowski M, Kaczmarczyk J, Kubaszewski Ł. Comparison of Bone Tissue Trace Element Content in the Different Radiological Stages of Hip Osteoarthritis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:3260. [PMID: 33809862 PMCID: PMC8004155 DOI: 10.3390/ijerph18063260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/14/2021] [Accepted: 03/20/2021] [Indexed: 01/02/2023]
Abstract
Bone metabolism and the trace element content associated with it change at each stage of degenerative disease. The aim of this study was to find out about the role of the analyzed elements in different stages of hip osteoarthritis. Elements associated with oxidative and enzymatic processes were analyzed depending on the changes in the radiological images of the hip joint. Element content analysis was performed by the inductively coupled plasma mass spectrometry analytical technique. The femoral head in severely osteoarthritic hips (KL3-4) compared to mild grade osteoarthritis (KL2) had a greater content of Cu (median 1.04 vs. 0.04), Sr (median 38.71 vs. 29.59), and Zn (median 75.12 vs. 63.21). There were no significant differences in the content of Mo, Cr, and Fe in the femoral head and neck between the groups. The Cu/Fe correlation was negative in the KL2 group (-0.47) and positive in the KL3-4 groups (0.45). Changes in the content and correlation of trace elements in the hip joint explain the changes in metabolism dependent on the severity of degenerative changes.
Collapse
Affiliation(s)
- Mikołaj Dąbrowski
- Adult Spine Orthopaedics Department, Poznan University of Medical Sciences, 61-545 Poznan, Poland;
| | - Anetta Zioła-Frankowska
- Department of Analytical Chemistry, Faculty of Chemistry, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland;
| | - Marcin Frankowski
- Department of Analytical and Environmental Chemistry, Faculty of Chemistry, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland;
| | - Jacek Kaczmarczyk
- Poznan LAB, Institute of Practical Medicine, 62-081 Przezmierowo, Poland;
| | - Łukasz Kubaszewski
- Adult Spine Orthopaedics Department, Poznan University of Medical Sciences, 61-545 Poznan, Poland;
| |
Collapse
|
16
|
Martini N, Parente JE, Restrepo-Guerrero G, Franca CA, Piro OE, Echeverría GA, Williams PA, Ferrer EG. Tetraacuo-bis-(N,N-dimethylacetamide-O)magnesium(II) chloride dihydrate. An option to improve magnesium effect on phosphatase stimulation and albumin binding. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
17
|
Abstract
In terms of antinociceptive action, the main mode of action of magnesium involves its antagonist action at the N-methyl-d-aspartate (NMDA) receptor, which prevents central sensitization and attenuates preexisting pain hypersensitivity. Given the pivotal function of NMDA receptors in pain transduction, magnesium has been investigated in a variety of pain conditions. The oral and parenteral administration of magnesium via the intravenous, intrathecal, or epidural route may alleviate pain and perioperative anesthetic and analgesic requirements. These beneficial effects of magnesium therapy have also been reported in patients with neuropathic pain, such as malignancy-related neurologic symptoms, diabetic neuropathy, postherpetic neuralgia, and chemotherapy-induced peripheral neuropathy. In addition, magnesium treatment is reportedly able to alleviate fibromyalgia, dysmenorrhea, headaches, and acute migraine attacks. Although magnesium plays an evolving role in pain management, better understanding of the mechanism underlying its antinociceptive action and additional clinical studies is required to clarify its role as an adjuvant analgesic.
Collapse
|
18
|
No YJ, Castilho M, Ramaswamy Y, Zreiqat H. Role of Biomaterials and Controlled Architecture on Tendon/Ligament Repair and Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904511. [PMID: 31814177 DOI: 10.1002/adma.201904511] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 11/10/2019] [Indexed: 06/10/2023]
Abstract
Engineering synthetic scaffolds to repair and regenerate ruptured native tendon and ligament (T/L) tissues is a significant engineering challenge due to the need to satisfy both the unique biological and biomechanical properties of these tissues. Long-term clinical outcomes of synthetic scaffolds relying solely on high uniaxial tensile strength are poor with high rates of implant rupture and synovitis. Ideal biomaterials for T/L repair and regeneration need to possess the appropriate biological and biomechanical properties necessary for the successful repair and regeneration of ruptured tendon and ligament tissues.
Collapse
Affiliation(s)
- Young Jung No
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Training Centre for Innovative BioEngineering, Sydney, NSW, 2006, Australia
| | - Miguel Castilho
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| | - Yogambha Ramaswamy
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Training Centre for Innovative BioEngineering, Sydney, NSW, 2006, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Training Centre for Innovative BioEngineering, Sydney, NSW, 2006, Australia
- Radcliffe Institute for Advanced Study, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
19
|
Singh S, Melnik R. Domain Heterogeneity in Radiofrequency Therapies for Pain Relief: A Computational Study with Coupled Models. Bioengineering (Basel) 2020; 7:E35. [PMID: 32272567 PMCID: PMC7355452 DOI: 10.3390/bioengineering7020035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 03/25/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
The objective of the current research work is to study the differences between the predicted ablation volume in homogeneous and heterogeneous models of typical radiofrequency (RF) procedures for pain relief. A three-dimensional computational domain comprising of the realistic anatomy of the target tissue was considered in the present study. A comparative analysis was conducted for three different scenarios: (a) a completely homogeneous domain comprising of only muscle tissue, (b) a heterogeneous domain comprising of nerve and muscle tissues, and (c) a heterogeneous domain comprising of bone, nerve and muscle tissues. Finite-element-based simulations were performed to compute the temperature and electrical field distribution during conventional RF procedures for treating pain, and exemplified here for the continuous case. The predicted results reveal that the consideration of heterogeneity within the computational domain results in distorted electric field distribution and leads to a significant reduction in the attained ablation volume during the continuous RF application for pain relief. The findings of this study could provide first-hand quantitative information to clinical practitioners about the impact of such heterogeneities on the efficacy of RF procedures, thereby assisting them in developing standardized optimal protocols for different cases of interest.
Collapse
Affiliation(s)
- Sundeep Singh
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, 75 University Avenue West, Waterloo, ON N2L 3C5, Canada;
| | - Roderick Melnik
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, 75 University Avenue West, Waterloo, ON N2L 3C5, Canada;
- BCAM—Basque Center for Applied Mathematics, Alameda de Mazarredo 14, E-48009 Bilbao, Spain
| |
Collapse
|
20
|
Singh S, Melnik R. Thermal ablation of biological tissues in disease treatment: A review of computational models and future directions. Electromagn Biol Med 2020; 39:49-88. [PMID: 32233691 DOI: 10.1080/15368378.2020.1741383] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Percutaneous thermal ablation has proven to be an effective modality for treating both benign and malignant tumours in various tissues. Among these modalities, radiofrequency ablation (RFA) is the most promising and widely adopted approach that has been extensively studied in the past decades. Microwave ablation (MWA) is a newly emerging modality that is gaining rapid momentum due to its capability of inducing rapid heating and attaining larger ablation volumes, and its lesser susceptibility to the heat sink effects as compared to RFA. Although the goal of both these therapies is to attain cell death in the target tissue by virtue of heating above 50°C, their underlying mechanism of action and principles greatly differs. Computational modelling is a powerful tool for studying the effect of electromagnetic interactions within the biological tissues and predicting the treatment outcomes during thermal ablative therapies. Such a priori estimation can assist the clinical practitioners during treatment planning with the goal of attaining successful tumour destruction and preservation of the surrounding healthy tissue and critical structures. This review provides current state-of-the-art developments and associated challenges in the computational modelling of thermal ablative techniques, viz., RFA and MWA, as well as touch upon several promising avenues in the modelling of laser ablation, nanoparticles assisted magnetic hyperthermia and non-invasive RFA. The application of RFA in pain relief has been extensively reviewed from modelling point of view. Additionally, future directions have also been provided to improve these models for their successful translation and integration into the hospital work flow.
Collapse
Affiliation(s)
- Sundeep Singh
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | - Roderick Melnik
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, Waterloo, Ontario, Canada.,BCAM - Basque Center for Applied Mathematics, Bilbao, Spain
| |
Collapse
|
21
|
Li Z, Peng Y, Pang X, Tang B. Potential Therapeutic Effects of Mg/HCOOH Metal Organic Framework on Relieving Osteoarthritis. ChemMedChem 2020; 15:13-16. [PMID: 31691487 DOI: 10.1002/cmdc.201900546] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/26/2019] [Indexed: 01/10/2023]
Abstract
This study was designed to explore the potential therapeutic effects of Mg-based metal organic frameworks (MOFs) in osteoarthritis (OA) and to broaden the application field of MOFs. Mg/HCOOH-MOF was introduced and characterized by using SEM, XRD, TGA, and FTIR. ICP-MS results proved that Mg/HCOOH-MOF is stable in various physiological media and can maintain the release of Mg2+ ions for a long time. In vitro experiments were carried out to evaluate its potential application in attenuating OA. Mg/HCOOH-MOF was successfully synthesized, and results of MTT assays showed that it is biocompatible and can promote cell proliferation. qPCR results suggested that it can significantly regulate the expression of OCN, Axin2, iNOS and IL-1β, which indicated its activity in anti-inflammation and bone protection. We believe that Mg/HCOOH-MOF could benefit OA therapy.
Collapse
Affiliation(s)
- Zhen Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.,Department of Physics and Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
| | - Ying Peng
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xiangchao Pang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.,Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, 518055, Guangdong, China.,Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| |
Collapse
|
22
|
Li S, Ma F, Pang X, Tang B, Lin L. Synthesis of chondroitin sulfate magnesium for osteoarthritis treatment. Carbohydr Polym 2019; 212:387-394. [DOI: 10.1016/j.carbpol.2019.02.061] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/05/2019] [Accepted: 02/18/2019] [Indexed: 01/27/2023]
|
23
|
Szychlinska MA, Di Rosa M, Castorina A, Mobasheri A, Musumeci G. A correlation between intestinal microbiota dysbiosis and osteoarthritis. Heliyon 2019; 5:e01134. [PMID: 30671561 PMCID: PMC6330556 DOI: 10.1016/j.heliyon.2019.e01134] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/21/2018] [Accepted: 01/09/2019] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative disease of the articular cartilage, resulting in pain and total joint disability. Recent studies focused on the role of the metabolic syndrome in inducing or worsening joint damage suggest that chronic low-grade systemic inflammation may represent a possible linking factor. This finding supports the concept of a new phenotype of OA, a metabolic OA. The gut microbiome is fundamental for human physiology and immune system development, among the other important functions. Manipulation of the gut microbiome is considered an important topic for the individual health in different medical fields such as medical biology, nutrition, sports, preventive and rehabilitative medicine. Since intestinal microbiota dysbiosis is strongly associated with the pathogenesis of several metabolic and inflammatory diseases, it is conceivable that also the pathogenesis of OA might be related to it. However, the mechanisms and the contribution of intestinal microbiota metabolites in OA pathogenesis are still not clear. The aim of this narrative review is to review recent literature concerning the possible contribution of dysbiosis to OA onset and to discuss the importance of gut microbiome homeostasis maintenance for optimal general health preservation.
Collapse
Affiliation(s)
- Marta Anna Szychlinska
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Alessandro Castorina
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
- Discipline of Anatomy & Histology, School of Medical Sciences, The University of Sydney, NSW, Australia
| | - Ali Mobasheri
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Arthritis Research UK Centre for Musculoskeletal Ageing Research, Queen's Medical Centre, Nottingham, UK
- Department of Regenerative Medicine, State Research Institute, Centre for Innovative Medicine, Lithuania
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
- School of the Sport of the Italian National Olympic Committee "CONI" Sicily, Italy
- Corresponding author.
| |
Collapse
|
24
|
Xu T, He X, Chen Z, He L, Lu M, Ge J, Weng J, Mu Y, Duan K. Effect of magnesium particle fraction on osteoinduction of hydroxyapatite sphere-based scaffolds. J Mater Chem B 2019; 7:5648-5660. [PMID: 31465084 DOI: 10.1039/c9tb01162e] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
HAs-30Mg (incorporation of 30% Mg into HA sphere-based scaffolds) induced the optimum new bone formation.
Collapse
Affiliation(s)
- Taotao Xu
- Key Lab of Advanced Technologies of Materials (MOE)
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | - Xu He
- Key Lab of Advanced Technologies of Materials (MOE)
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | - Zhenghui Chen
- Department of Stomatology
- Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital
- Chengdu
- China
| | - Lei He
- Key Lab of Advanced Technologies of Materials (MOE)
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | - Mengjie Lu
- Sichuan Provincial Lab of Orthopaedic Engineering
- Department of Bone and Joint Surgery
- Affiliated Hospital of Southwest Medical University
- Luzhou
- China
| | - Jianhua Ge
- Sichuan Provincial Lab of Orthopaedic Engineering
- Department of Bone and Joint Surgery
- Affiliated Hospital of Southwest Medical University
- Luzhou
- China
| | - Jie Weng
- Key Lab of Advanced Technologies of Materials (MOE)
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | - Yandong Mu
- Department of Stomatology
- Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital
- Chengdu
- China
| | - Ke Duan
- Sichuan Provincial Lab of Orthopaedic Engineering
- Department of Bone and Joint Surgery
- Affiliated Hospital of Southwest Medical University
- Luzhou
- China
| |
Collapse
|
25
|
Kosik-Bogacka DI, Lanocha-Arendarczyk N, Kot K, Zietek P, Karaczun M, Prokopowicz A, Kupnicka P, Ciosek Z. Calcium, magnesium, zinc and lead concentrations in the structures forming knee joint in patients with osteoarthritis. J Trace Elem Med Biol 2018; 50:409-414. [PMID: 30262313 DOI: 10.1016/j.jtemb.2018.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 07/10/2018] [Accepted: 08/28/2018] [Indexed: 12/29/2022]
Abstract
The aim of the study was to investigate the relationships between the concentrations of calcium (Ca), magnesium (Mg), zinc (Zn), and lead (Pb) in cartilage, anterior cruciate ligament, and meniscus samples obtained following knee joint surgery in patients with osteoarthritis in northwestern Poland. Furthermore, we examined the relationships between the concentrations of these metals in the studied parts of the knee joint and the influences of gender, age, BMI and hypertension. We found significantly higher concentrations of Ca, Mg, and Zn in the cartilage of men than in women and a significantly higher Pb concentration in the meniscus of the men. We also found a higher concentration of Pb in the cartilage of patients over 65 years of age. There were no differences in the concentrations of the studied metals between patients with and without hypertension. There was no relationship between Ca, Mg, Zn, and Pb levels in analyzed materials and BMI. Furthermore, we noted some new interactions between metals in the studied structures of the knee joint. The results reported in the study shows the influence of age, gender and BMI on the Ca, Mg, Zn and Pb in the studied structures of the knee joint.
Collapse
Affiliation(s)
- Danuta I Kosik-Bogacka
- Departament of Biology and Medical Parasitology, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland.
| | - Natalia Lanocha-Arendarczyk
- Departament of Biology and Medical Parasitology, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Karolina Kot
- Departament of Biology and Medical Parasitology, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Pawel Zietek
- Chair and Clinic of Orthopaedics, Traumatology and Oncology, Pomeranian Medical University, Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Maciej Karaczun
- Chair and Clinic of Orthopaedics, Traumatology and Oncology, Pomeranian Medical University, Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Adam Prokopowicz
- Departmet of Chemical Hazards and Genetic Toxicology, Institute of Occupational Medicina and Environmental Health, Koscielna 13, 71-200, Sosnowiec, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Zaneta Ciosek
- Departament of Biology and Medical Parasitology, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| |
Collapse
|
26
|
Chondroprotective Effects and Mechanisms of Dextromethorphan: Repurposing Antitussive Medication for Osteoarthritis Treatment. Int J Mol Sci 2018. [PMID: 29534535 PMCID: PMC5877686 DOI: 10.3390/ijms19030825] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disorder and primarily affects older people. The ideal anti-OA drug should have a modest anti-inflammatory effect and only limited or no toxicity for long-term use. Because the antitussive medication dextromethorphan (DXM) is protective in atherosclerosis and neurological diseases, two common disorders in aged people, we examined whether DXM can be protective in pro-inflammatory cytokine-stimulated chondrocytes and in a collagen-induced arthritis (CIA) animal model in this study. Chondrocytes were prepared from cartilage specimens taken from pigs or OA patients. Western blotting, quantitative PCR, and immunohistochemistry were adopted to measure the expression of collagen II (Col II) and matrix metalloproteinases (MMP). DXM significantly restored tumor necrosis factor-alpha (TNF-α)-mediated reduction of collagen II and decreased TNF-α-induced MMP-13 production. To inhibit the synthesis of MMP-13, DXM blocked TNF-α downstream signaling, including I kappa B kinase (IKK)α/β-IκBα-nuclear factor-kappaB (NF-κB) and c-Jun N-terminal kinase (JNK)-activator protein-1 (AP-1) activation. Besides this, DXM protected the CIA mice from severe inflammation and cartilage destruction. DXM seemed to protect cartilage from inflammation-mediated matrix degradation, which is an irreversible status in the disease progression of osteoarthritis. The results suggested that testing DXM as an osteoarthritis therapeutic should be a focus in further research.
Collapse
|
27
|
Hu T, Xu H, Wang C, Qin H, An Z. Magnesium enhances the chondrogenic differentiation of mesenchymal stem cells by inhibiting activated macrophage-induced inflammation. Sci Rep 2018; 8:3406. [PMID: 29467509 PMCID: PMC5821731 DOI: 10.1038/s41598-018-21783-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/09/2018] [Indexed: 12/22/2022] Open
Abstract
Magnesium deficiency increases the generation of pro-inflammatory cytokines, which is consistently accompanied by the sensitization of cells such as neutrophils, macrophages and endothelial cells. We investigated the potential of magnesium to regulate macrophage polarization and macrophage-induced inflammation with or without lipopolysaccharide (LPS) and interferon-γ (IFN-γ) activation and further elucidated whether these effects impact the inhibitory functions of activated macrophage-induced inflammation on cartilage regeneration. The results showed that magnesium inhibited the activation of macrophages, as indicated by a significant reduction in the percentage of CCR7-positive cells, while the percentage of CD206-positive cells decreased to a lesser degree. After activation, both pro-inflammatory and anti-inflammatory cytokines were down-regulated at the mRNA level and certain cytokines (IL-1β, IL-6 and IL-10) were decreased in the cell supernatant with the addition of magnesium. Moreover, magnesium decreased the nuclear translocation and phosphorylation of nuclear factor-κB (NF-κB) to impede its activation. A modified micromass culture system was applied to assess the effects of activated macrophage-conditioned medium with or without magnesium treatment on the chondrogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs). Magnesium enhanced the chondrogenic differentiation of hBMSCs by reversing the adverse effects of activated macrophage-induced inflammation.
Collapse
Affiliation(s)
- Tu Hu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Haitao Xu
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongyang Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hui Qin
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhiquan An
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
28
|
Diet-Intestinal Microbiota Axis in Osteoarthritis: A Possible Role. Mediators Inflamm 2016; 2016:3495173. [PMID: 27610004 PMCID: PMC5005536 DOI: 10.1155/2016/3495173] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 07/25/2016] [Indexed: 12/15/2022] Open
Abstract
Intestinal microbiota is highly involved in host physiology and pathology through activity of the microbiome and its metabolic products. Osteoarthritis (OA) is a common form of arthritis characterized by articular cartilage destruction and osteophyte formation. Although various person-level risk factors, such as age, sex, and obesity, have been proposed for the pathogenesis of OA, the underlying links between these person-level factors and OA are still enigmatic. Based on the current understanding in the crosstalk between intestinal microbiota and these risk factors, intestinal microbiota could be considered as a major hidden risk factor that provides a unifying mechanism to explain the involvement of these person-level risk factors in OA.
Collapse
|