1
|
Huang Y, Pan W, Bao H, Xu C, Ma J. Serum and synovial fluid levels of CSF-1 and in knee osteoarthritis and its clinical significance. BMC Musculoskelet Disord 2025; 26:353. [PMID: 40211184 PMCID: PMC11984276 DOI: 10.1186/s12891-025-08537-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 03/17/2025] [Indexed: 04/12/2025] Open
Abstract
OBJECTIVE To investigate the serum and synovial fluid levels of CSF-1 in patients with knee osteoarthritis (KOA) and evaluate its clinical significance. METHODS We selected 143 patients with KOA who received treatment at our hospital from June 2021 to August 2024. Enzyme-linked immunosorbent assay (ELISA) was used to determine the levels of CSF-1, IL-6, IL-1β, CRP, and HIF-1α in the serum of all study subjects, as well as the levels of these markers in the synovial fluid of all KOA patients. The Kellgren and Lawrence (KL) grading system was used to assess the radiographic severity of all KOA patients. Additionally, we also collected the Visual Analog Scale (VAS) scores and the Western Ontario McMaster University Osteoarthritis Index (WOMAC). Western blot (WB) was used to detect the expression levels of inflammatory factors in macrophages after CSF-1 stimulation. RESULTS Compared to healthy volunteers, KOA patients exhibited significantly elevated levels of serum CSF-1, IL-6, IL-1β, CRP, and HIF-1α (p < 0.05). The advanced group of KOA patients had significantly higher levels of serum and synovial fluid CSF-1 compared to the early group. Synovial fluid CSF-1 levels were associated with inflammation and disease severity in KOA patients. CSF-1 stimulation significantly increased the expression of CSF-1R, IL-6, TNF-α, IL-1β, HIF-1α, and MMP-3 in macrophages. Moreover, synovial fluid and serum CSF-1, synovial fluid HIF-1α, and synovial fluid IL-6 were identified as risk factors for advanced KOA. CONCLUSION Our findings indicated that the serum and synovial fluid levels of CSF-1 were significantly increased in KOA patients, even higher in patients with KL grade 3-4. Moreover, CSF-1 was identified as a risk factor associated with advanced stage KOA.
Collapse
Affiliation(s)
- Yuanchi Huang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, 555 East Friendship Road, South Gate, Xi'an, 710054, China
| | - Wenjie Pan
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, 555 East Friendship Road, South Gate, Xi'an, 710054, China
| | - Huanli Bao
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, 555 East Friendship Road, South Gate, Xi'an, 710054, China
| | - Chao Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, 555 East Friendship Road, South Gate, Xi'an, 710054, China.
| | - Jianbing Ma
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, 555 East Friendship Road, South Gate, Xi'an, 710054, China.
| |
Collapse
|
2
|
Li J, Zhang Y, Gan X, Li J, Xia G, He L, Xia C, Zhang W, Akhtar Ali K, Zhu M, Huang H. Blocking the LRH-1/LCN2 axis by ML-180, an LRH-1 inverse agonist, ameliorates osteoarthritis via inhibiting the MAPK pathway. Biochem Pharmacol 2025; 237:116922. [PMID: 40194607 DOI: 10.1016/j.bcp.2025.116922] [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: 11/17/2024] [Revised: 02/28/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
Osteoarthritis (OA) is a chronic and degenerative disease marked by inflammation and extracellular matrix (ECM) degeneration, contributing to synovial inflammation and cartilage destruction. Accumulating evidence has demonstrated that Liver receptor homolog-1 (LRH-1), an orphan nuclear receptor, mediates inflammatory response. However, there is a lack of evidence regarding the regulatory role of LRH-1 in OA pathogenesis. In this study, we confirmed that chondrocytes expressed LRH-1, and observed its upregulation in both IL-1β-treated chondrocytes and cartilage of destabilization of the medial meniscus (DMM)-operated mice. Overexpression of LRH-1 promoted inflammation and dysregulation of ECM metabolism in IL-1β-induced chondrocytes, reversed by inhibition of LRH-1 with ML-180 or gene silencing to protect chondrocytes. Moreover, ML-180 treatment in vivo improved the deteriorated OA phenotypes in mouse models, alleviating OA development. Mechanistically, RNA sequencing revealed that Lipocalin-2 (LCN2), a member of the lipocalin family associated with inflammation, is located downstream of LRH-1 and is positively regulated by it. Furthermore, the LRH-1/LCN2 axis mainly relied on activating the mitogen-activated protein kinase (MAPK) signaling pathway to promote inflammation and dysregulation of ECM metabolism, ultimately damaging chondrocytes. Our findings demonstrate that LRH-1 positively modulates LCN2,activating the MAPK pathway, indicating that targeting the LRH-1/LCN2/MAPK axis may represent a potential therapeutic strategy for OA.
Collapse
Affiliation(s)
- Jianwen Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yayun Zhang
- Department of Traumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xin Gan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Junhong Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ganqing Xia
- Department of Orthopedics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Lingxiao He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chengyan Xia
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weikai Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Khan Akhtar Ali
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Meipeng Zhu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Hui Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
3
|
Wu H, Li Y, Shi L, Liu Y, Shen J. New Advances in Periodontal Functional Materials Based on Antibacterial, Anti-Inflammatory, and Tissue Regeneration Strategies. Adv Healthc Mater 2025; 14:e2403206. [PMID: 39895157 DOI: 10.1002/adhm.202403206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/17/2025] [Indexed: 02/04/2025]
Abstract
With the global population aging, awareness of oral health is rising. Periodontitis, a widespread bacterial infectious disease, is gaining attention. Current novel biomaterials address key clinical issues like bacterial infection, gum inflammation, tooth loosening, and loss, focusing on antibacterial, anti-inflammatory, and tissue regeneration properties. However, strategies that integrate the advantages of these biomaterials to achieve synergistic therapeutic effects by clearing oral biofilms, inhibiting inflammation activation, and restoring periodontal soft and hard tissue functions remain very limited. Recent studies highlight the link between periodontitis and systemic diseases, underscoring the complexity of the periodontal disease. There is an urgent need to find comprehensive treatment plans that address clinical requirements. Whether by integrating new biomaterials to enhance existing periodontal treatments or by developing novel approaches to replace traditional therapies, these efforts will drive advancements in periodontitis treatment. Therefore, this review compares novel biomaterials with traditional treatments. It highlights the design concepts and mechanisms of these functional materials, focusing on their antibacterial, anti-inflammatory, and tissue regeneration properties, and discusses the importance of developing comprehensive treatment strategies. This review aims to provide guidance for emerging periodontitis research and to promote the development of precise and efficient treatment strategies.
Collapse
Affiliation(s)
- Haoyue Wu
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yuanfeng Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jing Shen
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| |
Collapse
|
4
|
Kim Y, Kim H, Yun SY, Lee BK. Primed IFN-γ-Umbilical Cord Stem Cells Ameliorate Temporomandibular Joint Osteoarthritis. Tissue Eng Part A 2025; 31:351-360. [PMID: 38787325 DOI: 10.1089/ten.tea.2023.0370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a degenerative disorder affecting the temporomandibular joint (TMJ), marked by persistent inflammation and structural damage to the joint. Only symptomatic treatment is available for managing TMJOA. Human umbilical cord mesenchymal stem cells (hUC-MSCs) show potential for treating TMJOA via their immune-modulating actions in the disease area. In addition, stimulation of inflammatory cytokines such as interferon-gamma in hUC-MSCs improves the therapeutic activity of naïve stem cells. Emerging evidence indicates that macrophages play significant roles in regulating joint inflammation through diverse secreted mediators in the pathogenesis of TMJOA. This study was conducted to evaluate the effects of inflammatory cytokine-stimulated hUC-MSCs in repairing TMJOA-induced cartilage lesions and the role of macrophages in the disease. Our in vitro data showed that stimulated hUC-MSCs induce M2 polarization of macrophages and enhance the expression of anti-inflammatory molecules. These effects were subsequently validated in vivo. In a rat model of TMJOA, stimulated hUC-MSCs ameliorated inflammation and increased M2 macrophages ratio. Our results indicate that hUC-MSCs stimulated by inflammatory cytokines modulate the activation of M2 macrophages, thereby shifting the local osteoarthritis microenvironment toward a prochondrogenic state and facilitating cartilage repair in inflammatory conditions. Stimulating hUC-MSCs with inflammatory cytokines could potentially offer an effective therapeutic approach for TMJOA, with macrophages playing a pivotal role in immune modulation.
Collapse
Affiliation(s)
- Yerin Kim
- AMIST, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Korea
| | - Hyunjeong Kim
- Asan Institute for Life Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, Seoul, Korea
| | - So-Yeon Yun
- AMIST, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Korea
| | - Bu-Kyu Lee
- AMIST, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Korea
- Asan Institute for Life Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, Seoul, Korea
- Department of Oral and Maxillofacial Surgery, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Korea
| |
Collapse
|
5
|
Zhou D, Luo Y, Li F, Liu T, Mei Y, Li F, Hou X, Fu Z, Liu Z. Exploring the mechanisms of PANoptosis in osteoarthritis and the therapeutic potential of andrographolide through bioinformatics and single-cell analysis. Biol Direct 2025; 20:41. [PMID: 40165317 PMCID: PMC11956211 DOI: 10.1186/s13062-025-00629-8] [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: 12/19/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease marked by the breakdown of cartilage, where apoptosis plays a key role. Although apoptosis-related genes in OA have been studied, a detailed analysis of PANoptosis-related genes and the search for therapeutic drugs remains limited. METHODS We performed a bioinformatics analysis combined with single-cell RNA sequencing to examine PANoptosis-related gene expression in OA cartilage. Key PANoptosis genes and critical cell populations involved in OA progression were identified. Drug prediction led to the selection of Andrographolide (AG), whose effects were validated through molecular docking, Western blotting, and qRT-PCR in chondrocyte models. RESULTS Several PANoptosis-related genes, including CASP8, TLR3, CASP1, and IL18, were significantly differentially expressed in OA. These genes are linked to processes such as apoptosis, pyroptosis, and the inflammasome complex. Pathway analysis revealed necroptosis, Toll-like receptor, and apoptosis signaling pathways as important in OA pathology. Single-cell analysis identified HomC, EC, and preHTC as key cell populations. AG was predicted to regulate PANoptosis genes, which was confirmed experimentally, demonstrating AG's potential to modulate key genes involved in cartilage degeneration. CONCLUSION This study highlights PANoptosis-related genes in OA and identifies Andrographolide as a promising therapeutic drug, offering new insights into OA treatment strategies.
Collapse
Affiliation(s)
- Daqian Zhou
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Yingjin Luo
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Fengjiang Li
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Tao Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Yongliang Mei
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Feilong Li
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Xianghan Hou
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Zhijiang Fu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
| | - Zongchao Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
- Luzhou Longmatan District People's Hospital, Luzhou, Sichuan, China.
| |
Collapse
|
6
|
Zou Z, Hu W, Kang F, Xu Z, Li Y, Zhang J, Li J, Zhang Y, Dong S. Interplay between lipid dysregulation and ferroptosis in chondrocytes and the targeted therapy effect of metformin on osteoarthritis. J Adv Res 2025; 69:515-529. [PMID: 38621621 PMCID: PMC11954841 DOI: 10.1016/j.jare.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/03/2024] [Accepted: 04/13/2024] [Indexed: 04/17/2024] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is a devastating whole-joint disease affecting a large population worldwide; the role of lipid dysregulation in OA and mechanisms underlying targeted therapy effect of lipid-lowering metformin on OA remains poorly defined. OBJECTIVES To investigate the effects of lipid dysregulation on OA progression and to explore lipid dysregulation-targeting OA treatment of metformin. METHODS RNA-Seq data, biochemical, and histochemical assays in human and murine OA cartilage as well as primary chondrocytes were utilized to determine lipid dysregulation. Effects of metformin, a potent lipid-lowering medication, on ACSL4 expression and chondrocyte metabolism were determined. Further molecular experiments, including RT-qPCR, western blotting, flow cytometry, and immunofluorescence staining, were performed to investigate underlying mechanisms. Mice with intra-articular injection of metformin were utilized to determine the effects on ACLT-induced OA progression. RESULTS ACSL4 and 4-HNE expressions were elevated in human and ACLT-induced mouse OA cartilage and IL-1β-treated chondrocytes (P < 0.05). Ferrostatin-1 largely rescued IL-1β-induced MDA, lipid peroxidation, and ferroptotic mitochondrial morphology (P < 0.05). Metformin decreased the levels of OA-related genes (P < 0.05) and increased the levels of p-AMPK and p-ACC in IL-1β-treated chondrocytes. Intra-articular injection of metformin alleviated ACLT-induced OA lesions in mice, and reverted the percentage of chondrocytes positive for MMP13, Col2a1, ACSL4 and 4-HNE in ACLT mice (P < 0.05). Ferroptotic chondrocytes promoted the recruitment and chemotaxis of RAW264.7 cells via CCL2, which was blocked by metformin in vitro (P < 0.05). CONCLUSION We establish a critical role of polyunsaturated fatty acids metabolic process in OA cartilage degradation and define metformin as a potential OA treatment. Metformin reshapes lipid availability and ameliorates chondrocyte ferroptosis sensitivity via the AMPK/ACC pathway. In the future, gene-edited animals and extensive omics technologies will be utilized to reveal detailed lipids' involvement in cartilage lesions.
Collapse
Affiliation(s)
- Zhi Zou
- College of Bioengineering, Chongqing University, Chongqing 400044, China; Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Kang
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhonghua Xu
- Joint Disease & Sport Medicine Center, Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yuheng Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing Zhang
- College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jianmei Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yuan Zhang
- Joint Disease & Sport Medicine Center, Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| | - Shiwu Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
7
|
Zhao F, Jia Z, Zhang L, Liu G, Li J, Zhao J, Xie Y, Chen L, Jiang H, He W, Wang A, Peng J, Zheng Y. A MnO 2 nanosheets doping double crosslinked hydrogel for cartilage defect repair through alleviating inflammation and guiding chondrogenic differentiation. Biomaterials 2025; 314:122875. [PMID: 39454507 DOI: 10.1016/j.biomaterials.2024.122875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/29/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024]
Abstract
The inflammatory microenvironment and inferior chondrogenesis are major symptoms after cartilage defect. Although various modifications strategies associated with hydrogels exhibit remarkable capacity of pro-cartilage regeneration, the adverse effect by prolonging inflammation is still formidable to hamper potential biomedical applications of different hydrogel implants. Herein, inspired by the repair microenvironment of articular cartilage defects, an injectable, immunomodulatory, and chondrogenic L-MNS-CMDA hydrogel is prepared through grafting vinyl and catechol groups to chitosan macromolecules using amide reaction, then further loading MnO2 nanosheets (MNS). The double crosslinking of photopolymerization and catechol oxidative polymerization endows L-MNS-CMDA hydrogel with preferable mechanical property, affording a suitable mechanical support for cartilage defect repair. Additionally, the robust tissue adhesion capability stemming from catechol groups guarantees the long-term retention of the hydrogel in the defect site. Meanwhile, L-MNS-CMDA hydrogel decomposes exogenous and intracellular H2O2 into O2 and H2O, to effectively alleviate cellular oxidative stress caused by long-term hypoxia. Under the synergies of catechol groups and MNS, L-MNS-CMDA hydrogel not only inhibits macrophages polarizing into M1 phenotype, but encourages them turn into M2 phenotype, thereby, reconstructing an immunization friendly microenvironment to ultimately enhance cartilage regeneration. Predictably, the hydrogel markedly induces rat bone marrow mesenchymal stem cells differentiating into chondrocytes by expressing abundant glycosaminoglycan and type II collagen. A cartilage defect model of rat knee joint indicates that L-MNS-CMDA hydrogel visually regulate the early inflammatory response of post-implantation, and facilitate cartilage regeneration and recovery of joint function after 12 weeks of post-implantation. All in all, this multifunctional L-MNS-CMDA hydrogel exhibits superior immunomodulatory and chondrogenic properties, holding immense clinical potential in the treatment of cartilage defects.
Collapse
Affiliation(s)
- Feilong Zhao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Zhibo Jia
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Liyang Zhang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Guodong Liu
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Junfei Li
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Jianming Zhao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Yajie Xie
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Lu Chen
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Hongyu Jiang
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei He
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Aiyuan Wang
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yudong Zheng
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| |
Collapse
|
8
|
Guo P, Alhaskawi A, Adel Abdo Moqbel S, Pan Z. Recent development of mitochondrial metabolism and dysfunction in osteoarthritis. Front Pharmacol 2025; 16:1538662. [PMID: 40017603 PMCID: PMC11865096 DOI: 10.3389/fphar.2025.1538662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Osteoarthritis is a degenerative joint disorder characterized by cartilage degradation, synovial inflammation, and altered subchondral bone structure. Recent insights have identified mitochondrial dysfunction as a pivotal factor in OA pathogenesis, contributing to chondrocyte apoptosis, oxidative stress, and extracellular matrix degradation. Disruptions in mitochondrial dynamics, including impaired biogenesis, mitophagy, and metabolic shifts from oxidative phosphorylation to glycolysis, exacerbate cartilage damage by promoting the production of reactive oxygen species and matrix-degrading enzymes such as ADAMTS and MMPs. This review explores the molecular mechanisms underlying mitochondrial dysfunction in OA, emphasizing its role in cartilage homeostasis and inflammation. Furthermore, it highlights emerging therapeutic strategies targeting mitochondrial pathways, including antioxidants, mitophagy enhancers, and metabolic modulators, as potential interventions to mitigate disease progression, which offer promising avenues for advancing personalized and disease-modifying treatments in OA.
Collapse
Affiliation(s)
- Pengchao Guo
- Emergency Department, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ahmad Alhaskawi
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Safwat Adel Abdo Moqbel
- Emergency Department, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijun Pan
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Xu H, He Y, Chen S, Meng C, Liu Q, Huang XJ, You HB. Blocking the CCL5/CCL7-CCR1 axis regulates macrophage polarization through NF-κB pathway to alleviate the progression of osteoarthritis. Int Immunopharmacol 2025; 147:114027. [PMID: 39805173 DOI: 10.1016/j.intimp.2025.114027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 12/21/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
OBJECTIVE To study the effect of CCR1 and its ligands on macrophage polarization and evaluate its effect on chondrocytes in relieving the progression of osteoarthritis. METHODS RAW cells were polarized to M1/M2 subtype, and then different concentrations of BX471 were added to selectively inhibit CCR1. The polarization of the cells was detected by RT-qPCR, immunofluorescence and flow cytometry. CCL5 and CCL7 genes were silenced by SiRNA and its role in macrophage polarization was analyzed. Macrophage conditioned medium was further used to stimulate chondrocytes. Histological observation was carried out on models of medial meniscus (DMM) with or without BX471 treatment. RESULTS We found that blocking of CCR1 and silencing of its ligand, CCL5 and CCL7, reduced the polarization of M1 macrophages. In terms of mechanism, we found that blocking CCR1 could reduce the activation of NF-κB pathway and inhibit the phosphorylation of IKK, IκBα and P65. In addition, blocking of CCR1 could also reduce cartilage injury induced by macrophage conditioned medium. In vivo, blocking of CCR1 reduced the infiltration and accumulation of M1 macrophages and alleviated articular cartilage injury. CONCLUSION CCL5/CCL7-CCR1 axis was involved in macrophage polarization, and blocking it could reduce synovitis and alleviate the process of OA.
Collapse
Affiliation(s)
- Hanqing Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, Hubei Province, China
| | - Yi He
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 1 Minde Road, Donghu, Nanchang 330006, Jiangxi, China
| | - Sheng Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, Hubei Province, China
| | - Chen Meng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, Hubei Province, China
| | - Qingyi Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, Hubei Province, China
| | - Xiao-Jian Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, Hubei Province, China.
| | - Hong-Bo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, Hubei Province, China.
| |
Collapse
|
10
|
Wang W, Chu Y, Lu Y, Xu J, Zhao W, Liang Z, Guo X, Xi L, Han T, Shen Y, Song W, Tang Y, Wen M, Qian Z, Wang L, Fan Z, Zhou G, Ren W. Skatole Alleviates Osteoarthritis by Reprogramming Macrophage Polarization and Protecting Chondrocytes. RESEARCH (WASHINGTON, D.C.) 2025; 8:0604. [PMID: 39902346 PMCID: PMC11788598 DOI: 10.34133/research.0604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/07/2025] [Accepted: 01/16/2025] [Indexed: 02/05/2025]
Abstract
Osteoarthritis (OA) is the most prevalent joint disease, yet effective disease-modifying OA drugs (DMOADs) remain elusive. Targeting macrophage polarization has emerged as a promising avenue for OA treatment. This study identified skatole through high-throughput screening as an efficient modulator of macrophage polarization. In vivo experiments demonstrated that skatole administration markedly reduced synovitis and cartilage damage in both destabilization of medial meniscus (DMM)-induced OA mice and monosodium iodoacetate (MIA)-induced OA rats. Mechanistically, skatole activated signal transducer and activator of transcription 6 (Stat6) signaling, promoting M2 macrophage polarization, while inhibiting nuclear factor-κB (NFκB) and mitogen-activated protein kinase (MAPK) signaling pathways to suppress M1 polarization. RNA-sequencing analysis, targeted metabolomics, and mitochondrial stress tests further revealed that skatole treatment shifted macrophages toward oxidative phosphorylation for energy production. Additionally, it up-regulated genes associated with glutathione metabolism and reactive oxygen species (ROS) pathways, reducing intracellular ROS production. The CUT&Tag assay results indicated that the downstream transcription factor p65 of NFκB can directly bind to gene loci related to inflammation, oxidative phosphorylation, and glutathione metabolism, thereby modulating gene expression. This regulatory process is inhibited by skatole. At the chondrocyte level, conditional medium from skatole-treated M1 macrophages balanced anabolism and catabolism in mouse chondrocytes and inhibited apoptosis. In IL1β-treated chondrocytes, skatole suppressed inflammation and catabolism without affecting apoptosis or anabolism. Overall, skatole maintains immune microenvironment homeostasis by modulating macrophage polarization in joints and preserves cartilage function by balancing chondrocyte anabolism and catabolism, effectively alleviating OA. These findings suggest skatole's potential as a DMOAD.
Collapse
Affiliation(s)
- Weiyun Wang
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
- The First Affiliated Hospital,
Xinxiang Medical University, Xinxiang 453199, China
| | - Yaru Chu
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Yunkun Lu
- Department of General Surgery, Sir Run Run Shaw Hospital,
Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Jie Xu
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Weixuan Zhao
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
- The First Affiliated Hospital,
Xinxiang Medical University, Xinxiang 453199, China
| | - Zhuo Liang
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Xueqiang Guo
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Lingling Xi
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Tao Han
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Yaping Shen
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Wenjuan Song
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Yanhua Tang
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Mengnan Wen
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Zhuang Qian
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Lei Wang
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Zhenlin Fan
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
| | - Guangdong Zhou
- Shanghai Key Lab of Tissue Engineering, Shanghai 9th People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wenjie Ren
- Institutes of Health Central Plain, Clinical Medical Center of Tissue Engineering and Regeneration,
Xinxiang Medical University, Xinxiang 453003, China
- The First Affiliated Hospital,
Xinxiang Medical University, Xinxiang 453199, China
| |
Collapse
|
11
|
Jin WS, Yin LX, Sun HQ, Zhao Z, Yan XF. Mesenchymal Stem Cells Injection Is More Effective Than Hyaluronic Acid Injection in the Treatment of Knee Osteoarthritis With Similar Safety: A Systematic Review and Meta-analysis. Arthroscopy 2025; 41:318-332. [PMID: 39154667 DOI: 10.1016/j.arthro.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024]
Abstract
PURPOSE To evaluate the efficacy and safety of intra-articular injection of mesenchymal stem cells (MSCs) versus hyaluronic acid (HA) in the treatment of knee osteoarthritis (KOA). METHODS Eligible randomized controlled trials (RCTs) were identified through a search of PubMed, Embase, the Cochrane Library, Web of Science, SinoMed, and CNKI databases from inception to March 2024. For meta-analysis, data on clinical outcomes were measured using visual analog scale (VAS) and the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), and data on cartilage repair were measured using the Whole-Organ Magnetic Resonance Imaging Score (WORMS); data on safety were evaluated by the incidence of adverse events. Two researchers independently read the included literature, extracted data and evaluated the quality, used the Cochrane risk bias assessment tool for bias risk assessment, and used RevMan5.3 software for meta-analysis. RESULTS Ten RCTs involving 818 patients with KOA ranging from I to Ⅲ on the Kellgren-Lawrence grading scale were included in this meta-analysis. Meta-analysis results showed that at 12 months, the WOMAC total score (mean difference [MD] = -10.22, 95% confidence interval [CI]: -14.86 to -5.59, P < .0001, Z = 4.32), VAS score (MD = -1.31, 95% CI: -1.90 to -0.73, P < .0001, Z = 4.40); and WORMS score (MD = -26.01, 95% CI: -31.88 to -20.14, P < .001, Z = 8.69) of the MSCs group all decreased significantly (P < .05) compared with the HA control group and reached the minimal clinically important differences. Furthermore, there was no significant difference in the incidence of adverse events (relative risk = 1.54, 95% CI: 0.85-2.79, P = .16, I2 = 0) between the 2 groups (P > .05). CONCLUSIONS In terms of efficacy, the clinical effects of intra-articular injection therapy using MSCs for KOA are superior to those of HA, and the cartilage repair effect of MSCs is also markedly better than that of HA. Although the clinical effects varied across time periods, the functional score reached the minimum clinically significant difference at both 6 and 12 months. In terms of safety, adverse reactions mainly manifest as joint pain, swelling, and joint effusion. Both intra-articular injections of MSCs and HA did not result in severe adverse reactions, indicating that MSCs and HA have similar safety profiles. LEVEL OF EVIDENCE Level I, meta-analysis of Level I studies.
Collapse
Affiliation(s)
- Wen-Shu Jin
- Department of Hospital-Acquired Infection Control, The Second Affiliated Hospital of Shandong First Medical University, Taian City, Shandong Province, China
| | - Lu-Xu Yin
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan City, Shandong Province, China
| | - Hua-Qiang Sun
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan City, Shandong Province, China
| | - Zhang Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan City, Shandong Province, China
| | - Xin-Feng Yan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan City, Shandong Province, China.
| |
Collapse
|
12
|
Li Z, Li X, Xia H, Wang Y, Wei N. NEK2 promotes the progression of osteoarthritis by stabilizing ATF2 through phosphorylation at Ser-112 and inhibiting autophagy. Int Immunopharmacol 2025; 146:113833. [PMID: 39693952 DOI: 10.1016/j.intimp.2024.113833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/27/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024]
Abstract
NEK2 (NIMA-related kinase 2) has recently gained attention for its potential role in osteoarthritis (OA) chondrocytes, however, its specific involvement remains unclear. This study aimed to investigate the role of NEK2 in OA progression and the underlying molecular mechanisms. Primary mouse knee chondrocytes were stimulated with IL-1β to establish an in vitro OA model, followed by the knockdown of NEK2 or ATF2. The results indicated that silencing NEK2 or ATF2 impeded the IL-1β-induced decrease in cell proliferation and increase in inflammation, extracellular matrix (ECM) degradation, and apoptosis in chondrocytes. NEK2 or ATF2 knockdown restored IL-1β-induced autophagy defects. Mechanistically, NEK2 interacts with ATF2 to reduce its ubiquitylation level and enhance its stability by phosphorylating ATF2 at Ser-112. Consistently, ATF2 overexpression reversed the protective effect of NEK2 silencing on IL-1β-induced autophagy defects and chondrocyte injury. Additionally, a mouse OA model was established using medial meniscus destabilization (DMM) surgery, and NEK2 was knocked down by intra-articular injection of an adenovirus-mediated NEK2 interference vector. Downregulation of NEK2 mitigated cartilage degradation andautophagy defects ina mouse OA model. In conclusion, NEK2 promoted OA progression by enhancing ATF2 stability by phosphorylating it at Ser-112.
Collapse
Affiliation(s)
- Zhiqin Li
- Rheumatology and Immunology Department, Xi'an Third Hospital, Xi'an, China
| | - Xiaofeng Li
- Cardiovascular Surgery, Xianyang First People's Hospital, Xianyang, China
| | - Hongli Xia
- Rheumatology and Immunology Department, Xi'an Third Hospital, Xi'an, China
| | - Yiqi Wang
- Rheumatology and Immunology Department, Xi'an Third Hospital, Xi'an, China
| | - Ning Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.
| |
Collapse
|
13
|
Maurer S, Kirsch V, Ruths L, Brenner RE, Riegger J. Senolytic therapy combining Dasatinib and Quercetin restores the chondrogenic phenotype of human osteoarthritic chondrocytes by the release of pro-anabolic mediators. Aging Cell 2025; 24:e14361. [PMID: 39402753 PMCID: PMC11995296 DOI: 10.1111/acel.14361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/21/2024] [Accepted: 09/13/2024] [Indexed: 01/11/2025] Open
Abstract
Cellular senescence is associated with various age-related disorders and is assumed to play a major role in the pathogenesis of osteoarthritis (OA). Based on this, we tested a senolytic combination therapy using Dasatinib (D) and Quercetin (Q) on aged isolated human articular chondrocytes (hACs), as well as in OA-affected cartilage tissue (OARSI grade 1-2). Stimulation with D + Q selectively eliminated senescent cells in both, cartilage explants and isolated hAC. Furthermore, the therapy significantly promoted chondroanabolism, as demonstrated by increased gene expression levels of COL2A1, ACAN, and SOX9, as well as elevated collagen type II and glycosaminoglycan biosynthesis. Additionally, D + Q treatment significantly reduced the release of SASP factors (IL6, CXCL1). RNA sequencing analysis revealed an upregulation of the anabolic factors, inter alia, FGF18, IGF1, and TGFB2, as well as inhibitory effects on cytokines and the YAP-1 signaling pathway, explaining the underlying mechanism of the chondroanabolic promotion upon senolytic treatment. Accordingly, stimulation of untreated hAC with conditioned medium of D + Q-treated cells similarly induced the expression of chondrogenic markers. Detailed analyses demonstrated that chondroanabolic effects could be mainly attributed to Dasatinib, while monotherapeutical application of Quercetin or Navitoclax did not promote the chondroanabolism. Overall, D + Q therapy restored the chondrogenic phenotype in OA hAC most likely by creating a pro-chondroanabolic environment through the reduction of SASP factors and upregulation of growth factors. This senolytic approach could therefore be a promising candidate for further testing as a disease-modifying osteoarthritis drug.
Collapse
Affiliation(s)
- Svenja Maurer
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of OrthopedicsUniversity of UlmUlmGermany
| | - Valeria Kirsch
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of OrthopedicsUniversity of UlmUlmGermany
| | - Leonie Ruths
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of OrthopedicsUniversity of UlmUlmGermany
| | - Rolf E. Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of OrthopedicsUniversity of UlmUlmGermany
| | - Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of OrthopedicsUniversity of UlmUlmGermany
| |
Collapse
|
14
|
Yu H, Ding C, Hu Z, Liu Q, Gu X, He J, Yan Y, Yu S, Gao L, Cheng W, Wu Z, Jing J. Biocompatible ionized air alleviates rat osteoarthritis by modulating polarization from M1 to M2 macrophages. Sci Rep 2024; 14:31901. [PMID: 39738316 PMCID: PMC11685818 DOI: 10.1038/s41598-024-83198-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 12/12/2024] [Indexed: 01/02/2025] Open
Abstract
The imbalance in the proportion of M1/M2 macrophage polarization is a crucial contributor to the persistent progression of osteoarthritis (OA). This study aimed to evaluate the effects of low-dose biocompatible ionized air (BIA) on macrophage polarization and its subsequent chondroprotective effects, thereby validating the potential of BIA in slowing the progression of OA. In vitro experiments demonstrated that BIA modulates the polarization of M1 macrophages toward the M2 phenotype via the ROS-mediated STAT6 pathway. This shift reduces the expression of pro-inflammatory mediators while increasing the expression of anti-inflammatory mediators and pro-chondrogenic factors, leading to an improved microenvironment surrounding chondrocytes. The direct benefits of this improved microenvironment include enhanced chondrocyte viability, inhibition of apoptosis, and reduced degradation of the extracellular matrix. In vivo studies in rats showed that BIA inhibited M1 macrophage infiltration in the synovium, upregulated the proportion of M2 macrophages, alleviated cartilage degeneration, and delayed OA progression. This gas-based regulatory strategy may open new avenues for the treatment of OA.
Collapse
Affiliation(s)
- Haoran Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
| | - Chengbiao Ding
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
| | - Zhongyao Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
| | - Qi Liu
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei, 230000, China
| | - Xuesong Gu
- The Second Clinical Medical College of Anhui Medical University, Hefei, 230000, China
| | - Junyan He
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
| | - Yiqun Yan
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
| | - Shenrui Yu
- The Second Clinical Medical College of Anhui Medical University, Hefei, 230000, China
| | - Lin Gao
- The First Clinical Medical College of Anhui Medical University, Hefei, 230000, China
| | - Wendan Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China.
| | - Zhengwei Wu
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei, 230000, China.
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China.
| |
Collapse
|
15
|
Liu Y, Zhang Z, Fang Y, Liu C, Zhang H. Ferroptosis in Osteoarthritis: Current Understanding. J Inflamm Res 2024; 17:8471-8486. [PMID: 39529997 PMCID: PMC11552513 DOI: 10.2147/jir.s493001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease in elderly people that is characterized by cartilage loss and abrasion, leading to joint pain and dysfunction. The aetiology of OA is complicated and includes abnormal mechanical stress, a mild inflammatory environment, chondrocyte senescence and apoptosis, and changes in chondrocyte metabolism. Ferroptosis is a regulated cell death modality characterized by the excessive accumulation of lipid peroxidation and mitochondrial dysfunction. The role of ferroptosis in OA pathogenesis has aroused researchers' attention in the past two years, and there is mounting evidence indicating that ferroptosis is destructive. However, the impact of ferroptosis on OA and how the regulators of ferroptosis affect OA development are unclear. Here, we reviewed the current understanding of ferroptosis in OA pathogenesis and summarized several drugs and compounds targeting ferroptosis in OA treatment. The accumulation of intracellular iron, the trigger of Fenton reaction, the excessive production of ROS, the peroxidation of PUFA-PLs, and mitochondrial and membrane damage are involved in chondrocyte ferroptosis. System Xc - and GPX4 are the most important regulators that control ferroptosis. Several compounds, such as DFO and Fer-1, have been proven effective in preventing ferroptosis and slowing OA progression on animal models. Collectively, targeting ferroptosis shows great potential in treating OA.
Collapse
Affiliation(s)
- Yikai Liu
- Department of Orthopaedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People’s Republic of China
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Zian Zhang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Yuan Fang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Chang Liu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Haining Zhang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| |
Collapse
|
16
|
Pemmari A, Moilanen E. Macrophage and chondrocyte phenotypes in inflammation. Basic Clin Pharmacol Toxicol 2024; 135:537-549. [PMID: 39319534 DOI: 10.1111/bcpt.14085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024]
Abstract
Inflammation is a complex biological process protecting the body from diverse external threats. Effectively performing this task requires an intricate, well-regulated interplay of different cells and tissues. Furthermore, several cells participating in inflammation can assume diverse phenotypes. A classic and relatively well-studied example of phenotypic diversity in inflammation is macrophage polarization. Based on the TH1/TH2 phenotypes of T helper cells, this scheme has proinflammatory "classical/M1" activation contrasted with the anti-inflammatory and healing-promoting "alternative/M2" phenotype. Some authors have extended the concept into an M17 phenotype induced by the classic TH17 cytokine IL-17. Phenotypic changes in chondrocytes have also been studied especially in the context of osteoarthritis (OA), and there are indications that these cells can also assume polarized phenotypes at least partly analogous to those of TH cells and macrophages. The therapeutic success of biological agents targeting TH1/TH2/TH17 inductor and/or effector cytokines displays the utility of the concept of polarization. The aim of this focused review is to survey the internal and external factors affecting macrophage and chondrocyte phenotypes (such as inflammatory cytokines, widely used medications and natural products) and to explore the possibility of ameliorating pathological states by modulating these phenotypes.
Collapse
Affiliation(s)
- Antti Pemmari
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
17
|
Liu H, Jiang L, Xu S, Wang C, Sun J. Quercetin prevents methylmercury-induced mitochondrial dysfunction in the cerebral cortex of mice. Drug Chem Toxicol 2024; 47:1124-1138. [PMID: 38647114 DOI: 10.1080/01480545.2024.2341888] [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: 08/26/2023] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
Methylmercury (MeHg) exposure can cause nerve damage and mitochondrial dysfunction. Mitochondrial dysfunction is mainly mediated by mitochondrial biogenesis and mitochondrial dynamics disorders. Quercetin (QE) plays an important role in activating silencing information regulator 2 related enzyme 1 (SIRT1), and SIRT1 activates peroxisome-proliferator-activated receptor-γ co-activator 1α (PGC-1α), which can regulate mitochondrial biogenesis and mitochondrial dynamics. The main purpose of this study was to explore the alleviating effects of QE on MeHg-induced nerve damage and mitochondrial dysfunction. The results showed that QE could reduce the excessive production of reactive oxygen species (ROS) and the loss of membrane potential induced by MeHg. Meanwhile, QE activated SIRT1 activity and SIRT1/PGC-1α signaling pathway, improved mitochondrial biogenesis and fusion and reduced mitochondrial fission. In summary, we hypothesized that QE prevents MeHg-induced mitochondrial dysfunction by activating SIRT1/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Haihui Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Liujiangshan Jiang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Si Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Chen Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Jingyi Sun
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, PR China
| |
Collapse
|
18
|
Liao J, Gu Q, Liu Z, Wang H, Yang X, Yan R, Zhang X, Song S, Wen L, Wang Y. Edge advances in nanodrug therapies for osteoarthritis treatment. Front Pharmacol 2024; 15:1402825. [PMID: 39539625 PMCID: PMC11559267 DOI: 10.3389/fphar.2024.1402825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
As global population and lifestyles change, osteoarthritis (OA) is becoming a major healthcare challenge world. OA, a chronic condition characterized by inflammatory and degeneration, often present with joint pain and can lead to irreversible disability. While there is currently no cure for OA, it is commonly managed using nonsteroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, and glucosamine. Although these treatments can alleviate symptoms, it is difficult to effectively deliver and sustain therapeutic agents within joints. The emergence of nanotechnology, particularly in form of smart nanomedicine, has introduced innovative therapeutic approaches for OA treatment. Nanotherapeutic strategies offer promising advantages, including more precise targeting of affected areas, prolonged therapeutic effects, enhanced bioavailability, and reduced systemic toxicity compared to traditional treatments. While nanoparticles show potential as a viable delivery system for OA therapies based on encouraging lab-based and clinical trials results, there remails a considerable gap between current research and clinical application. This review highlights recent advances in nanotherapy for OA and explore future pathways to refine and optimize OA treatments strategies.
Collapse
Affiliation(s)
- Jinfeng Liao
- Department of Dermatology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Qingjia Gu
- Department of ENT, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Zheng Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Xian Yang
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongkai Yan
- Department of Radiology, Ohio state university, Columbus, OH, United States
| | - Xiaofeng Zhang
- Greenwich Hospital, Yale New Haven Health, Greenwich, CT, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Lebin Wen
- Department of Thyroid, Sichuan Second Hospital of TCM, Chengdu, China
| | - Yi Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
19
|
Li Y, Gong H, Gan T, Ma X, Geng Q, Yin S, Zhang H, Wu Y. Smart Hydrogel Dressing Enhances the Healing of Chronic Infectious Diabetic Wounds through Dual-Barrier Drug Delivery Action. Biomacromolecules 2024; 25:6814-6829. [PMID: 39235955 DOI: 10.1021/acs.biomac.4c01041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Chronic diabetic wounds struggle to heal due to drug-resistant bacterial infections, oxidative stress microenvironment, and immune dysfunction. At present, the disease has become a huge clinical challenge. Multifunctional hydrogels with antibacterial, antioxidant, and anti-inflammatory properties are becoming an emerging trend in the treatment of chronic wounds. However, matching different bioactive functions with the wound healing process to sequentially exert antibacterial, antioxidant, anti-inflammatory, and immunomodulatory functions remains a significant challenge. In this research, a hydrogel dressing with bactericidal and anti-inflammatory properties was synthesized by crafting a pH/ROS-responsive scaffold from phenylboronic acid-grafted hyaluronic acid (HA-PBA) and 4-arm-PEG-dopamine (4A-PEG-Dopa), employing dynamic borate ester bonds. This structure was then infused with the antimicrobial peptide (AMP) and ROS-sensitive micelle mPEG-TK-PLGA loaded with quercetin (QC). This dressing embodied a dual-barrier drug delivery mechanism, engineered for the prolonged and consistent liberation of QC. In the experiment, the hydrogel dissociated within the acidic microenvironment of diabetic wounds, thereby liberating the encapsulated micelles and AMP. Upon further dissociation, the micelles release QC due to the ROS-abundant microenvironment, which could relieve oxidative stress and encourage M2 polarization of macrophage via the Akt/STAT6 signaling pathway. Therefore, this smart delivery system, developed through our innovative approach, holds promise for treating chronic infectious diabetic wounds.
Collapse
Affiliation(s)
- Yaxing Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Heng Gong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tingjiang Gan
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xikun Ma
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qirui Geng
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shijiu Yin
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hui Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ye Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
20
|
Ilyas S, Baek CY, Manan A, Choi Y, Jo HG, Lee D. Mechanistic Exploration of Smilax glabra Roxb. in Osteoarthritis: Insights from Network Pharmacology, Molecular Docking, and In Vitro Validation. Pharmaceuticals (Basel) 2024; 17:1285. [PMID: 39458926 PMCID: PMC11510151 DOI: 10.3390/ph17101285] [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: 08/27/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Arthritis, a debilitating joint disease, remains a significant global health burden. This study uncovers the therapeutic potential of the medicinal plant Smilax glabra Roxb. (SGR) in attenuating progression of disease by modulating immune responses. Methods: Through computational approaches, key bioactive compounds in SGR were identified by using freely available databases: TCMSP, TCMID, HIT2.0, HERB, and INPUT in order to elucidate their underlying mechanisms of action. Therapeutic targets for the disease have been retrieved by TTD, GeneCard, and OMIM databases. The STRING database was used to analyze the protein-protein interactions (PPI) of intersecting genes. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to reveal the functional roles of genes. Mcule was used for molecular docking and binding affinity of compounds and targets were evaluated by DeepPurpose model. ALP activity, cell viability assay, TRAP staining were also performed. Results: A total of 14 active SGR compounds with 59 common targets for arthritis have been identified. These targets have a major role in controlling biological processes such as wound healing, oxygen responses, and chemical stimuli. Molecular docking by Mcule platform demonstrated that quercetin and β-sitosterol showed higher binding energy affinities with TNF, TP53, PTGS2, and JUN as compared to other targets. To explore the complex relationship between compounds and targets, pre-trained Davis and KIBA models were used to predict the affinity values of selected compounds. In MC3T3-E1 cells, ALP activity was significantly increased and bone marrow macrophages (BMM) showed a low number of TRAP-positive cells in SGR-treated cells. Conclusions: Our findings demonstrate that SGR effectively inhibits/regulates inflammatory responses, prevents cartilage degradation, promotes bone regeneration, and can be used as a promising candidate for the development of novel arthritis treatment.
Collapse
Affiliation(s)
- Sidra Ilyas
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Chae Yun Baek
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Abdul Manan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Yeojin Choi
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Hee-Geun Jo
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
- Naturalis Inc. 6, Daewangpangyo-ro, Bundang-gu, Seongnam-si 13549, Republic of Korea
| | - Donghun Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| |
Collapse
|
21
|
Ruan X, Jin X, Sun F, Pi J, Jinghu Y, Lin X, Zhang N, Chen G. IGF signaling pathway in bone and cartilage development, homeostasis, and disease. FASEB J 2024; 38:e70031. [PMID: 39206513 DOI: 10.1096/fj.202401298r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The skeleton plays a fundamental role in the maintenance of organ function and daily activities. The insulin-like growth factor (IGF) family is a group of polypeptide substances with a pronounced role in osteoblast differentiation, bone development, and metabolism. Disturbance of the IGFs and the IGF signaling pathway is inextricably linked with assorted developmental defects, growth irregularities, and jeopardized skeletal structure. Recent findings have illustrated the significance of the action of the IGF signaling pathway via growth factors and receptors and its interactions with dissimilar signaling pathways (Wnt/β-catenin, BMP, TGF-β, and Hh/PTH signaling pathways) in promoting the growth, survival, and differentiation of osteoblasts. IGF signaling also exhibits profound influences on cartilage and bone development and skeletal homeostasis via versatile cell-cell interactions in an autocrine, paracrine, and endocrine manner systemically and locally. Our review summarizes the role and regulatory function as well as a potentially integrated gene network of the IGF signaling pathway with other signaling pathways in bone and cartilage development and skeletal homeostasis, which in turn provides an enlightening insight into visualizing bright molecular targets to be eligible for designing effective drugs to handle bone diseases and maladies, such as osteoporosis, osteoarthritis, and dwarfism.
Collapse
Affiliation(s)
- Xinyi Ruan
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiuhui Jin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fuju Sun
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiashun Pi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yihan Jinghu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xinyi Lin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nenghua Zhang
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
22
|
Yu Q, Xiao Y, Guan M, Zhang X, Yu J, Han M, Li Z. Copper metabolism in osteoarthritis and its relation to oxidative stress and ferroptosis in chondrocytes. Front Mol Biosci 2024; 11:1472492. [PMID: 39329090 PMCID: PMC11425083 DOI: 10.3389/fmolb.2024.1472492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Ferroptosis, an iron-ion-dependent process of lipid peroxidation, damages the plasma membrane, leading to non-programmed cell death. Osteoarthritis (OA), a prevalent chronic degenerative joint disease among middle-aged and older adults, is characterized by chondrocyte damage or loss. Emerging evidence indicates that chondrocyte ferroptosis plays a role in OA development. However, most research has concentrated on ferroptosis regulation involving typical iron ions, potentially neglecting the significance of elevated copper ions in both serum and joint fluid of patients with OA. This review aims to fill this gap by systematically examining the interplay between copper metabolism, oxidative stress, ferroptosis, and copper-associated cell death in OA. It will provide a comprehensive overview of copper ions' role in regulating ferroptosis and their dual role in OA. This approach seeks to offer new insights for further research, prevention, and treatment of OA.
Collapse
Affiliation(s)
- Qingyuan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yanan Xiao
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mengqi Guan
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xianshuai Zhang
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jianan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mingze Han
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Zhenhua Li
- Orthopedic Center, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
23
|
Xu D, Bi S, Li J, Ma S, Yu ZA, Wang Y, Chen H, Zhan J, Song X, Cai Y. Legumain-Guided Ferulate-Peptide Self-Assembly Enhances Macrophage-Endotheliocyte Partnership to Promote Therapeutic Angiogenesis After Myocardial Infarction. Adv Healthc Mater 2024:e2402056. [PMID: 39252665 DOI: 10.1002/adhm.202402056] [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: 06/24/2024] [Revised: 08/31/2024] [Indexed: 09/11/2024]
Abstract
Promoting angiogenesis and modulating the inflammatory microenvironment are promising strategies for treating acute myocardial infarction (MI). Macrophages are crucial in regulating inflammation and influencing angiogenesis through interactions with endothelial cells. However, current therapies lack a comprehensive assessment of pathological and physiological subtleties, resulting in limited myocardial recovery. In this study, legumain-guided ferulate-peptide nanofibers (LFPN) are developed to facilitate the interaction between macrophages and endothelial cells in the MI lesion and modulate their functions. LFPN exhibits enhanced ferulic acid (FA) aggregation and release, promoting angiogenesis and alleviating inflammation. The multifunctional role of LFPN is validated in cells and an MI mouse model, where it modulated macrophage polarization, attenuated inflammatory responses, and induces endothelial cell neovascularization compare to FA alone. LFPN supports the preservation of border zone cardiomyocytes by regulating inflammatory infiltration in the ischemic core, leading to significant functional recovery of the left ventricle. These findings suggest that synergistic therapy exploiting multicellular interaction and enzyme guidance may enhance the clinical translation potential of smart-responsive drug delivery systems to treat MI. This work emphasizes macrophage-endothelial cell partnerships as a novel paradigm to enhance cell interactions, control inflammation, and promote therapeutic angiogenesis.
Collapse
Affiliation(s)
- Delong Xu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Shenghui Bi
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jiejing Li
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Shaodan Ma
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Ze-An Yu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yenan Wang
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Huiming Chen
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jie Zhan
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xudong Song
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yanbin Cai
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
24
|
Miteva D, Kitanova M, Velikova T. Biomacromolecules as Immunomodulators: Utilizing Nature’s Tools for Immune Regulation. MACROMOL 2024; 4:610-633. [DOI: 10.3390/macromol4030037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Although there are numerous available immunomodulators, those of natural origin would be preferable based on their safety profile and effectiveness. The research and clinical interest in immunomodulators have increased in the last decades, especially in the immunomodulatory properties of plant-based therapies. Innovative technologies and extensive study on immunomodulatory natural products, botanicals, extracts, and active moieties with immunomodulatory potential could provide us with valuable entities to develop as novel immunomodulatory medicines to enhance current chemotherapies. This review focuses on plant-based immunomodulatory drugs that are currently in clinical studies. However, further studies in this area are of utmost importance to obtain complete information about the positive effects of medicinal plants and their chemical components and molecules as an alternative to combatting various diseases and/or prevention.
Collapse
Affiliation(s)
- Dimitrina Miteva
- Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tzankov 8 blv., 1164 Sofia, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, 1407 Sofia, Bulgaria
| | - Meglena Kitanova
- Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tzankov 8 blv., 1164 Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1407 Sofia, Bulgaria
| |
Collapse
|
25
|
Villagrán-Andrade KM, Núñez-Carro C, Blanco FJ, de Andrés MC. Nutritional Epigenomics: Bioactive Dietary Compounds in the Epigenetic Regulation of Osteoarthritis. Pharmaceuticals (Basel) 2024; 17:1148. [PMID: 39338311 PMCID: PMC11434976 DOI: 10.3390/ph17091148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Nutritional epigenomics is exceptionally important because it describes the complex interactions among food compounds and epigenome modifications. Phytonutrients or bioactive compounds, which are secondary metabolites of plants, can protect against osteoarthritis by suppressing the expression of inflammatory and catabolic mediators, modulating epigenetic changes in DNA methylation, and the histone or chromatin remodelling of key inflammatory genes and noncoding RNAs. The combination of natural epigenetic modulators is crucial because of their additive and synergistic effects, safety and therapeutic efficacy, and lower adverse effects than conventional pharmacology in the treatment of osteoarthritis. In this review, we have summarized the chondroprotective properties of bioactive compounds used for the management, treatment, or prevention of osteoarthritis in both human and animal studies. However, further research is needed into bioactive compounds used as epigenetic modulators in osteoarthritis, in order to determine their potential value for future clinical applications in osteoarthritic patients as well as their relation with the genomic and nutritional environment, in order to personalize food and nutrition together with disease prevention.
Collapse
Affiliation(s)
- Karla Mariuxi Villagrán-Andrade
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Carmen Núñez-Carro
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Francisco J Blanco
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
- Grupo de Investigación en Reumatología y Salud, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - María C de Andrés
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| |
Collapse
|
26
|
Falvino A, Gasperini B, Cariati I, Bonanni R, Chiavoghilefu A, Gasbarra E, Botta A, Tancredi V, Tarantino U. Cellular Senescence: The Driving Force of Musculoskeletal Diseases. Biomedicines 2024; 12:1948. [PMID: 39335461 PMCID: PMC11429507 DOI: 10.3390/biomedicines12091948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
The aging of the world population is closely associated with an increased prevalence of musculoskeletal disorders, such as osteoporosis, sarcopenia, and osteoarthritis, due to common genetic, endocrine, and mechanical risk factors. These conditions are characterized by degeneration of bone, muscle, and cartilage tissue, resulting in an increased risk of fractures and reduced mobility. Importantly, a crucial role in the pathophysiology of these diseases has been proposed for cellular senescence, a state of irreversible cell cycle arrest induced by factors such as DNA damage, telomere shortening, and mitochondrial dysfunction. In addition, senescent cells secrete pro-inflammatory molecules, called senescence-associated secretory phenotype (SASP), which can alter tissue homeostasis and promote disease progression. Undoubtedly, targeting senescent cells and their secretory profiles could promote the development of integrated strategies, including regular exercise and a balanced diet or the use of senolytics and senomorphs, to improve the quality of life of the aging population. Therefore, our review aimed to highlight the role of cellular senescence in age-related musculoskeletal diseases, summarizing the main underlying mechanisms and potential anti-senescence strategies for the treatment of osteoporosis, sarcopenia, and osteoarthritis.
Collapse
Affiliation(s)
- Angela Falvino
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Beatrice Gasperini
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Roberto Bonanni
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Angela Chiavoghilefu
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
| | - Elena Gasbarra
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
| | - Annalisa Botta
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Umberto Tarantino
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Department of Clinical Sciences and Translational Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
27
|
Kan T, Tian Z, Sun L, Kong W, Yan R, Yu Z, Tian QW, Liu C. Quercetin-Loaded Zeolitic Imidazolate Framework-8 (ZIF-8) Nanoparticles Attenuate Osteoarthritis by Activating Autophagy via the Pi3k/Akt Signaling. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40444-40454. [PMID: 39058329 DOI: 10.1021/acsami.4c04649] [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: 07/28/2024]
Abstract
Osteoarthritis treatment remains a significant clinical challenge. Quercetin, a natural flavonoid with anti-inflammatory and antiapoptotic properties, might be utilized to treat OA. However, poor water solubility and short joint retention duration limit its bioavailability and translation to clinical applications. A one-step self-assembly method was utilized to fabricate quercetin-loaded zeolitic imidazolate framework-8 (Qu@ZIF-8) nanoparticles using zinc ions, 2-methylimidazole, and quercetin. In vitro tests showed that Qu@ZIF-8 nanoparticles released pH-responsive agents into chondrocytes, effectively protecting them from interleukin (IL)-induced inflammation and apoptosis, thereby promoting cartilage anabolic activities. These underlying mechanisms revealed a remarkable increase of autophagy in IL-β-treated chondrocytes, followed by the inhibition of the Pi3k/Akt signaling pathway, which contributed to the protective effect of Qu @ZIF-8. By the establishment of medial meniscus instability (DMM) in OA mice, Qu@ZIF-8 substantially improved cartilage structural integrity and chondrocyte status, as well as attenuated OA progression. Importantly, Qu@ZIF-8 outperformed quercetin alone in the treatment of OA due to its control release. The combined research findings indicate that Qu@ZIF-8 shields chondrocytes from inflammation and apoptosis by activating autophagy and repressing the Pi3k/Akt pathway. This investigation may provide new insights for clinically extending the therapy of OA.
Collapse
Affiliation(s)
- Tianyou Kan
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| | - Zhengtao Tian
- The State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200100, China
| | - Lin Sun
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| | - Wei Kong
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| | - Ruisi Yan
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences Institution, Shanghai 200100 China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| | - Qi-Wei Tian
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences Institution, Shanghai 200100 China
| | - Chenglei Liu
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| |
Collapse
|
28
|
Zhu H, Cai C, Yu Y, Zhou Y, Yang S, Hu Y, Zhu Y, Zhou J, Zhao J, Ma H, Chen Y, Xu Y. Quercetin-Loaded Bioglass Injectable Hydrogel Promotes m6A Alteration of Per1 to Alleviate Oxidative Stress for Periodontal Bone Defects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403412. [PMID: 38749005 PMCID: PMC11304245 DOI: 10.1002/advs.202403412] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/21/2024] [Indexed: 08/09/2024]
Abstract
Periodontal disease ranks third among noncommunicable illnesses, behind cancer and cardiovascular disease, and is closely related to the occurrence and progression of various systemic diseases. However, elucidating the processes of periodontal disease and promoting periodontal bone regeneration remains a challenge. Here, quercetin is demonstrated to reduce the oxidative stress state of orofacial mesenchymal stem cells (OMSCs) in vitro and to affect the osteogenic growth of OMSCs through molecular mechanisms that mediate the m6A change in Per1. Nevertheless, the limited therapeutic efficacy of systemic medication and the limitations of local medication resulting from the small, moist, and highly dynamic periodontal environment make it challenging to treat periodontal tissues with medication. Herein, a biosafe injectable hydrogel drug-controlled delivery system is constructed as a bone-enhancing factory and loaded with quercetin to treat oxidative stress injury in periodontal tissues. This drug-carrying system made up of nanoscale bioglass microspheres and a light-cured injectable hydrogel, allows effective drug particle loading and cementation in the dynamic and moist periodontal environment. Furthermore, the system demonstrates the ability to stimulate OMSCs osteogenic differentiation in a Per1-dependent manner, which ultimately promotes periodontal bone repair, suggesting that this system has potential for clinical periodontal therapy.
Collapse
Affiliation(s)
- Huimin Zhu
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityNo. 639Zhizaoju RdShanghai200011China
| | - Chao Cai
- Zhejiang Engineering Research Center for Tissue Repair MaterialsWenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000China
| | - Yeke Yu
- Department of Oral Maxillofacial‑Head and Neck OncologyShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineNo 639, Zhizaoju RdShanghai200011China
| | - Yuning Zhou
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityNo. 639Zhizaoju RdShanghai200011China
| | - Shiyuan Yang
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityNo. 639Zhizaoju RdShanghai200011China
| | - Yue Hu
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityNo. 639Zhizaoju RdShanghai200011China
| | - Yan Zhu
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityNo. 639Zhizaoju RdShanghai200011China
| | - Jia Zhou
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityNo. 639Zhizaoju RdShanghai200011China
| | - Jieyun Zhao
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityNo. 639Zhizaoju RdShanghai200011China
| | - Hailong Ma
- Department of Oral Maxillofacial‑Head and Neck OncologyShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineNo 639, Zhizaoju RdShanghai200011China
| | - Yujie Chen
- State Key Laboratory of Metal Matrix CompositesSchool of Materials Science and EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Yuanjin Xu
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityNo. 639Zhizaoju RdShanghai200011China
| |
Collapse
|
29
|
Chen X, Liu J, Wang G, Sun Y, Ding X, Zhang X. Validation of Jianpi Qingre Tongluo Recipe in Reducing Inflammation and Dyslipidemia in Osteoarthritis via Lnc RNA HOTAIR/APN/PI3K/AKT. Int J Gen Med 2024; 17:3293-3318. [PMID: 39081673 PMCID: PMC11288358 DOI: 10.2147/ijgm.s466148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/20/2024] [Indexed: 08/02/2024] Open
Abstract
Objective Jianpi Qingre Tongluo Recipe (JQP) has been widely used in clinical practice, and its anti-Osteoarthritis (OA) effectiveness and specific mechanism have been concerned. This study aims to explore the clinical effect of JQP in reducing inflammation and dyslipidemia in OA and the molecular mechanism. Methods The clinical efficacy of JQP in OA treatment was assessed through data mining. Through the network pharmacology technology, the interactive network of "active component-target-disease" was developed, the interaction relationship of the related proteins was analyzed, and enrichment analysis of gene pathway biological process was conducted. Molecular docking was carried out with PyMOL and AutodockTools-1.5.7. Finally, cell experiments were used to verify JQP's delay of immune inflammation in OA. Results We found that JQP could ameliorate the immune inflammatory and lipid metabolism indicators; reduce VAS and SAS score in OA. A total of 98 genes overlapped between target genes of JQP and OA. TNF, IL-6, IL-1β, and AKT1 shared the highest centrality among all target genes. KEGG analysis unveiled that 98 intersection genes were predominantly enriched in PI3K/AKT pathway in the anti-OA system. In vitro, after peripheral blood mononuclear cell (PBMC) stimulation, inflammatory cytokines imbalances and the expressions of adiponectin (APN) were decreased in osteoarthritis-chondrocytes (OA-CH). Furthermore, JQP-containing serum protected OA-CHs through down-regulating HOTAIR levels, thereby up-regulating APN and depressing PI3K/AKT pathway. Conclusion This study suggests that JQP might reduce inflammation and improve lipid metabolism of OA by regulating HOTAIR/APN/PI3K/AKT. Our results bring a new solution for OA.
Collapse
Affiliation(s)
- Xiaolu Chen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, 230038, People’s Republic of China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230012, People’s Republic of China
- Anhui University of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Jian Liu
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, 230038, People’s Republic of China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230012, People’s Republic of China
| | - Guizhen Wang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, 230038, People’s Republic of China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230012, People’s Republic of China
| | - Yanqiu Sun
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, 230038, People’s Republic of China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230012, People’s Republic of China
| | - Xiang Ding
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, 230038, People’s Republic of China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230012, People’s Republic of China
- Anhui University of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Xianheng Zhang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, 230038, People’s Republic of China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230012, People’s Republic of China
- Anhui University of Traditional Chinese Medicine, Hefei, People’s Republic of China
| |
Collapse
|
30
|
Ruan H, Zhu T, Wang T, Guo Y, Liu Y, Zheng J. Quercetin Modulates Ferroptosis via the SIRT1/Nrf-2/HO-1 Pathway and Attenuates Cartilage Destruction in an Osteoarthritis Rat Model. Int J Mol Sci 2024; 25:7461. [PMID: 39000568 PMCID: PMC11242395 DOI: 10.3390/ijms25137461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, causing symptoms such as joint pain, swelling, and deformity, which severely affect patients' quality of life. Despite advances in medical treatment, OA management remains challenging, necessitating the development of safe and effective drugs. Quercetin (QUE), a natural flavonoid widely found in fruits and vegetables, shows promise due to its broad range of pharmacological effects, particularly in various degenerative diseases. However, its role in preventing OA progression and its underlying mechanisms remain unclear. In this study, we demonstrated that QUE has a protective effect against OA development both in vivo and in vitro, and we elucidated the underlying molecular mechanisms. In vitro, QUE inhibited the expression of IL-1β-induced chondrocyte matrix metalloproteinases (MMP3 and MMP13) and inflammatory mediators such as INOS and COX-2. It also promoted the expression of collagen II, thereby preventing the extracellular matrix (ECM). Mechanistically, QUE exerts its protective effect on chondrocytes by activating the SIRT1/Nrf-2/HO-1 and inhibiting chondrocyte ferroptosis. Similarly, in an OA rat model induced by anterior cruciate ligament transection (ACLT), QUE treatment improved articular cartilage damage, reduced joint pain, and normalized abnormal subchondral bone remodeling. QUE also reduced serum IL-1β, TNF-α, MMP3, CTX-II, and COMP, thereby slowing the progression of OA. QUE exerts chondroprotective effects by inhibiting chondrocyte oxidative damage and ferroptosis through the SIRT1/Nrf-2/HO-1 pathway, effectively alleviating OA progression in rats.
Collapse
Affiliation(s)
- Hongri Ruan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.R.); (T.Z.); (T.W.); (Y.G.)
| | - Tingting Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.R.); (T.Z.); (T.W.); (Y.G.)
| | - Tiantian Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.R.); (T.Z.); (T.W.); (Y.G.)
| | - Yingchao Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.R.); (T.Z.); (T.W.); (Y.G.)
| | - Yun Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.R.); (T.Z.); (T.W.); (Y.G.)
| | - Jiasan Zheng
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163000, China
| |
Collapse
|
31
|
Dada T, Mahalingam K, Bhartiya S. Reversing Aging and Improving Health Span in Glaucoma Patients: The Next Frontier? J Curr Glaucoma Pract 2024; 18:87-93. [PMID: 39575133 PMCID: PMC11576344 DOI: 10.5005/jp-journals-10078-1451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024] Open
Abstract
How to cite this article: Dada T, Mahalingam K, Bhartiya S. Reversing Aging and Improving Health Span in Glaucoma Patients: The Next Frontier? J Curr Glaucoma Pract 2024;18(3):87-93.
Collapse
Affiliation(s)
- Tanuj Dada
- Department of Ophthalmology, Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi, India
| | - Karthikeyan Mahalingam
- Department of Ophthalmology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Shibal Bhartiya
- Department of Ophthalmology and Community Outreach, Marengo Asia Hospitals, Gurugram and Faridabad, Haryana, India; Mayo Clinic, Jacksonville, Florida, United States
| |
Collapse
|
32
|
Meng X, Li Y, Wang F, Li T, Wang B, Wang Q, Long J, Xie H, Zhang Y, Li J. Quercetin attenuates inflammation in rosacea by directly targeting p65 and ICAM-1. Life Sci 2024; 347:122675. [PMID: 38688383 DOI: 10.1016/j.lfs.2024.122675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/08/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
AIMS Rosacea is an inflammatory skin disease with immune and vascular dysfunction. Although there are multiple treatment strategies for rosacea, the clinical outcomes are unsatisfactory. MAIN METHODS Combining transcriptome data and the Connectivity Map database quercetin was identified as a novel candidate for rosacea. Next, the therapeutic efficacy of quercetin was substantiated through proteomic analyses, in vivo experiments, and in vitro assays. Additionally, the utilization of DARTS, molecular docking and experimental verification revealed the therapeutic mechanisms of quercetin. KEY FINDINGS Treatment with quercetin resulted in the following effects: (i) it effectively ameliorated rosacea-like features by reducing immune infiltration and angiogenesis; (ii) it suppressed the expression of inflammatory mediators in HaCaT cells and HDMECs; (iii) it interacted with p65 and ICAM-1 directly, and this interaction resulted in the repression of NF-κB signal and ICAM-1 expression in rosacea. SIGNIFICANCE We show for the first time that quercetin interacted with p65 and ICAM-1 directly to alleviated inflammatory and vascular dysfunction, suggesting quercetin is a novel, promising therapeutic candidate for rosacea.
Collapse
Affiliation(s)
- Xin Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Yangfan Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Fan Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Ben Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Wang
- Hunan Binsis Biotechnology Co., Ltd, Changsha, China
| | - Juan Long
- Department of Dermatology, Hunan Children's Hospital, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Yiya Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
33
|
Hu D, Li R, Li Y, Wang M, Wang L, Wang S, Cheng H, Zhang Q, Fu C, Qian Z, Wei Q. Inflammation-Targeted Nanomedicines Alleviate Oxidative Stress and Reprogram Macrophages Polarization for Myocardial Infarction Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308910. [PMID: 38582507 DOI: 10.1002/advs.202308910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/27/2024] [Indexed: 04/08/2024]
Abstract
Myocardial infarction (MI) is a critical global health challenge, with current treatments limited by the complex MI microenvironment, particularly the excessive oxidative stress and intense inflammatory responses that exacerbate cardiac dysfunction and MI progression. Herein, a mannan-based nanomedicine, Que@MOF/Man, is developed to target the inflammatory infarcted heart and deliver the antioxidative and anti-inflammatory agent quercetin (Que), thereby facilitating a beneficial myocardial microenvironment for cardiac repair. The presence of mannan on the nanoparticle surface enables selective internalization by macrophages rather than cardiomyocytes. Que@MOF/Man effectively neutralizes reactive oxygen species in macrophages to reduce oxidative stress and promote their differentiation into a reparative phenotype, reconciling the inflammatory response and enhancing cardiomyocyte survival through intercellular communication. Owing to the recruitment of macrophages into inflamed myocardium post-MI, in vivo, administration of Que@MOF/Man in MI rats revealed the specific distribution into the injured myocardium compared to free Que. Furthermore, Que@MOF/Man exhibited favorable results in resolving inflammation and protecting cardiomyocytes, thereby preventing further myocardial remodeling and improving cardiac function in MI rats. These findings collectively validate the rational design of an inflammation-targeted delivery strategy to mitigate oxidative stress and modulate the inflammation response in the injured heart, presenting a therapeutic avenue for MI treatment.
Collapse
Affiliation(s)
- Danrong Hu
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ran Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yicong Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Meng Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Shiqi Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Hongxin Cheng
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Qing Zhang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Chenying Fu
- National Clinical Research Center for Geriatrics, Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Zhiyong Qian
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine in Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
34
|
Qi Z, Zhu J, Cai W, Lou C, Li Z. The role and intervention of mitochondrial metabolism in osteoarthritis. Mol Cell Biochem 2024; 479:1513-1524. [PMID: 37486450 PMCID: PMC11224101 DOI: 10.1007/s11010-023-04818-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/15/2023] [Indexed: 07/25/2023]
Abstract
Osteoarthritis (OA), a prevalent degenerative joint disease, affects a substantial global population. Despite the elusive etiology of OA, recent investigations have implicated mitochondrial dysfunction as a significant factor in disease pathogenesis. Mitochondria, pivotal cellular organelles accountable for energy production, exert essential roles in cellular metabolism. Hence, mitochondrial dysfunction can exert broad-ranging effects on various cellular processes implicated in OA development. This comprehensive review aims to provide an overview of the metabolic alterations occurring in OA and elucidate the diverse mechanisms through which mitochondrial dysfunction can contribute to OA pathogenesis. These mechanisms encompass heightened oxidative stress and inflammation, perturbed chondrocyte metabolism, and compromised autophagy. Furthermore, this review will explore potential interventions targeting mitochondrial metabolism as means to impede or decelerate the progression of OA. In summary, this review offers a comprehensive understanding of the involvement of mitochondrial metabolism in OA and underscores prospective intervention strategies.
Collapse
Affiliation(s)
- Zhanhai Qi
- Department of Orthopedics, The 960th hospital of the Joint Logistics Support Force of the People's Liberation Army, Jinan, Shandong, China
| | - Jiaping Zhu
- Department of Orthopedics, Jinan City People's Hospital, Jinan, Shandong, China
| | - Wusheng Cai
- Department of Orthopedics, Heze Third People's Hospital, Heze, Shandong, China
| | - Chunbiao Lou
- Department of Orthopedics, Heze Third People's Hospital, Heze, Shandong, China
| | - Zongyu Li
- Department of Orthopedics, The 960th hospital of the Joint Logistics Support Force of the People's Liberation Army, Jinan, Shandong, China.
| |
Collapse
|
35
|
El Oirdi M. Harnessing the Power of Polyphenols: A New Frontier in Disease Prevention and Therapy. Pharmaceuticals (Basel) 2024; 17:692. [PMID: 38931359 PMCID: PMC11206774 DOI: 10.3390/ph17060692] [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: 04/28/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
There are a wide variety of phytochemicals collectively known as polyphenols. Their structural diversity results in a broad range of characteristics and biological effects. Polyphenols can be found in a variety of foods and drinks, including fruits, cereals, tea, and coffee. Studies both in vitro and in vivo, as well as clinical trials, have shown that they possess potent antioxidant activities, numerous therapeutic effects, and health advantages. Dietary polyphenols have demonstrated the potential to prevent many health problems, including obesity, atherosclerosis, high blood sugar, diabetes, hypertension, cancer, and neurological diseases. In this paper, the protective effects of polyphenols and the mechanisms behind them are investigated in detail, citing the most recent available literature. This review aims to provide a comprehensive overview of the current knowledge on the role of polyphenols in preventing and managing chronic diseases. The cited publications are derived from in vitro, in vivo, and human-based studies and clinical trials. A more complete understanding of these naturally occurring metabolites will pave the way for the development of novel polyphenol-rich diet and drug development programs. This, in turn, provides further evidence of their health benefits.
Collapse
Affiliation(s)
- Mohamed El Oirdi
- Department of Life Sciences, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia;
- Department of Basic Sciences, Preparatory Year, King Faisal University, Al Ahsa 31982, Saudi Arabia
| |
Collapse
|
36
|
Lee YT, Mohd Yunus MH, Yazid MD, Ugusman A. Unraveling the path to osteoarthritis management: targeting chondrocyte apoptosis for therapeutic intervention. Front Cell Dev Biol 2024; 12:1347126. [PMID: 38827524 PMCID: PMC11140145 DOI: 10.3389/fcell.2024.1347126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/06/2024] [Indexed: 06/04/2024] Open
Abstract
Osteoarthritis (OA) is a chronic disease affecting joints and further causing disabilities. This disease affects around 240 million people worldwide. It is a multifactorial disease, and its etiology is difficult to determine. Although numerous therapeutic strategies are available, the therapies are aimed at reducing pain and improving patients' quality of life. Hence, there is an urgent need to develop disease-modifying drugs (DMOAD) that can reverse or halt OA progression. Apoptosis is a cell removal process that is important in maintaining homeostatic mechanisms in the development and sustaining cell population. The apoptosis of chondrocytes is believed to play an important role in OA progression due to poor chondrocytes self-repair abilities to maintain the extracellular matrix (ECM). Hence, targeting chondrocyte apoptosis can be one of the potential therapeutic strategies in OA management. There are various mediators and targets available to inhibit apoptosis such as autophagy, endoplasmic reticulum (ER) stress, oxidative stress, and inflammation. As such, this review highlights the importance and potential targets that can be aimed to reduce chondrocyte apoptosis.
Collapse
Affiliation(s)
- Yi Ting Lee
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Mohd Heikal Mohd Yunus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| |
Collapse
|
37
|
Coppola C, Greco M, Munir A, Musarò D, Quarta S, Massaro M, Lionetto MG, Maffia M. Osteoarthritis: Insights into Diagnosis, Pathophysiology, Therapeutic Avenues, and the Potential of Natural Extracts. Curr Issues Mol Biol 2024; 46:4063-4105. [PMID: 38785519 PMCID: PMC11119992 DOI: 10.3390/cimb46050251] [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: 02/28/2024] [Revised: 04/05/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Osteoarthritis (OA) stands as a prevalent and progressively debilitating clinical condition globally, impacting joint structures and leading to their gradual deterioration through inflammatory mechanisms. While both non-modifiable and modifiable factors contribute to its onset, numerous aspects of OA pathophysiology remain elusive despite considerable research strides. Presently, diagnosis heavily relies on clinician expertise and meticulous differential diagnosis to exclude other joint-affecting conditions. Therapeutic approaches for OA predominantly focus on patient education for self-management alongside tailored exercise regimens, often complemented by various pharmacological interventions primarily targeting pain alleviation. However, pharmacological treatments typically exhibit short-term efficacy and local and/or systemic side effects, with prosthetic surgery being the ultimate resolution in severe cases. Thus, exploring the potential integration or substitution of conventional drug therapies with natural compounds and extracts emerges as a promising frontier in enhancing OA management. These alternatives offer improved safety profiles and possess the potential to target specific dysregulated pathways implicated in OA pathogenesis, thereby presenting a holistic approach to address the condition's complexities.
Collapse
Affiliation(s)
- Chiara Coppola
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Via Lecce-Arnesano, 73100 Lecce, Italy; (C.C.); (A.M.)
| | - Marco Greco
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Anas Munir
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Via Lecce-Arnesano, 73100 Lecce, Italy; (C.C.); (A.M.)
| | - Debora Musarò
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Stefano Quarta
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy;
| | - Maria Giulia Lionetto
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Michele Maffia
- Department of Experimental Medicine, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| |
Collapse
|
38
|
Li B, Shen E, Wu Z, Qi H, Wu C, Liu D, Jiang X. BMSC-Derived Exosomes Attenuate Rat Osteoarthritis by Regulating Macrophage Polarization through PINK1/Parkin Signaling Pathway. Cartilage 2024:19476035241245805. [PMID: 38641989 PMCID: PMC11569690 DOI: 10.1177/19476035241245805] [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: 12/26/2023] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 04/21/2024] Open
Abstract
OBJECTIVE Exosomes derived from bone marrow mesenchymal stem cells (BMSC-Exos) may modulate the M1/M2 polarization of macrophages during osteoarthritis (OA). However, the underlying mechanisms of BMSC-Exos in this process still need to be elucidated. In this study, we explored the role of BMSC-Exos in the polarization of macrophages in vitro and the OA rats in vivo. METHODS The effects of BMSC-Exos on RAW264.7 cells were determined, including the production of reactive oxygen species (ROS) and the protein expression of Akt, PINK1, and Parkin. We prepared an OA model by resecting the anterior cruciate ligament and medial meniscus of Sprague-Dawley (SD) rats. Hematoxylin-eosin (H&E) and safranin O-fast green staining, immunohistochemistry and immunofluorescence analyses, and the examination of interleukin 6 (IL-6), interleukin 1β (IL-1β), tumor necrosis factor alpha (TNF-α), and interleukin 10 (IL-10) were performed to assess changes in cartilage and synovium. RESULTS BMSC-Exos inhibited mitochondrial membrane damage, ROS production, and the protein expression of PINK1 and Parkin. Akt phosphorylation was downregulated under lipopolysaccharide (LPS) induction but significantly recovered after treatment with BMSC-Exos. BMSC-Exos alleviated cartilage damage, inhibited M1 polarization, and promoted M2 polarization in the synovium in OA rats. The expression of PINK1 and Parkin in the synovium and the levels of IL-6, IL-1β, and TNF-α in the serum decreased, but the level of IL-10 increased when BMSC-Exos were used in OA rats. CONCLUSION BMSC-Exos ameliorate OA development by regulating synovial macrophage polarization, and one of the underlying mechanisms may be through inhibiting PINK1/Parkin signaling.
Collapse
Affiliation(s)
- Beibei Li
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Enpu Shen
- Shanghai Putuo District Central Hospital, Shanghai, China
| | - Zhiwen Wu
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Hui Qi
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Cheng’ai Wu
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Danping Liu
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xu Jiang
- Department of Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
39
|
Zhang J, Cheng F, Rong G, Tang Z, Gui B. IDN5706 Inhibits Synovial Inflammation via Inducing M2 Polarization of Synovial Macrophages in Osteoarthritis Rats. Pharmacology 2024; 109:156-168. [PMID: 38565085 DOI: 10.1159/000538452] [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: 12/07/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION IDN5706 is a tetrahydro derivative of hyperforin. In this study, we aimed to explore the effect of IDN5706 on synovial macrophages in osteoarthritis (OA) rats and the underlying mechanisms. METHODS OA rats were employed for the in vivo experiments, and RAW264.7 cells were employed for the in vitro experiments. Histopathological changes in synovium were examined using hematoxylin-eosin staining. Cell apoptosis in synovium was assessed by TUNEL staining. Macrophage polarization was determined by immunohistochemical analysis and flow cytometry. The mRNA expression and protein level of genes were detected by qRT-PCR and Western blot. The efferocytosis of macrophages was assessed by flow cytometry. RESULTS IDN5706 reversed the increased CD86-positive cells (M1 macrophages) and decreased CD206-positive cells (M2 macrophages), both in synovium and synovial fluid of OA rats. The in vitro experiments further confirmed the promotion effect of IDN5706 on M2 macrophages, accompanied by the elevated Arg-1 and reduced iNOS. Also, the upregulated p-mTOR in synovium and synovial fluid of OA rats were reversed by IDN5706, and the decreased M1 macrophages and increased M2 macrophages induced by IDN5706 were reversed by the mTOR activator. IDN5706 enhanced the efferocytosis of IL-4-treated RAW264.7 cells, and the animal experiments further revealed the involvement of efferocytosis in the improvement of OA by IDN5706. CONCLUSIONS IDN5706 enhanced the efferocytosis of synovial macrophages by inducing M2 polarization via inhibiting p-mTOR, thus suppressing synovial inflammation and OA development, providing a theoretical basis for IDN5706 as a clinical drug for inflammatory diseases.
Collapse
Affiliation(s)
- Jinling Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fangyue Cheng
- Department of Rheumatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Genxiang Rong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhi Tang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Binjie Gui
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
40
|
Kim H, Kim Y, Yun SY, Lee BK. Efficacy of IFN-γ-Primed Umbilical Cord-Derived Mesenchymal Stem Cells on Temporomandibular Joint Osteoarthritis. Tissue Eng Regen Med 2024; 21:473-486. [PMID: 38190096 PMCID: PMC10987468 DOI: 10.1007/s13770-023-00620-2] [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: 11/03/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Temporomandibular joint osteoarthritis (TMJOA) is a degenerative disease affecting the cartilage and subchondral bone, leading to temporomandibular joint pain and dysfunction. The complex nature of TMJOA warrants effective alternative treatments, and mesenchymal stem cells (MSCs) have shown promise in regenerative therapies. The aim of this study is twofold: firstly, to ascertain the optimal interferon-gamma (IFN-γ)-primed MSC cell line for TMJOA treatment, and secondly, to comprehensively evaluate the therapeutic efficacy of IFN-γ-primed mesenchymal stem cells derived from the human umbilical cord matrix in a rat model of TMJOA. METHODS We analyzed changes in the expression of several key genes associated with OA protection in MSC-secreted compounds. Following this, we performed co-culture experiments using a transwell system to predict gene expression changes in primed MSCs in the TMJOA environment. Subsequently, we investigated the efficacy of the selected IFN-γ-primed human umbilical cord matrix-derived MSCs (hUCM-MSCs) for TMJOA treatment in a rat model. RESULTS IFN-γ-primed MSCs exhibited enhanced expression of IDO, TSG-6, and FGF-2. Moreover, co-culturing with rat OA chondrocytes induced a decrease in pro-inflammatory and extracellular matrix degradation factors. In the rat TMJOA model, IFN-γ-primed MSCs with elevated IDO1, TSG-6, and FGF2 expression exhibited robust anti-inflammatory and therapeutic capacities, promoting the improvement of the inflammatory environment and cartilage regeneration. CONCLUSION These findings underscore the importance of prioritizing the mitigation of the inflammatory milieu in TMJOA treatment and highlight IFN-γ-primed MSCs secreting these three factors as a promising, comprehensive therapeutic strategy.
Collapse
Affiliation(s)
- Hyunjeong Kim
- Biomedical Engineering Research Center, Asan Medical Center, Asan Institute for Life Sciences, Seoul, Korea
| | - Yerin Kim
- Asan Medical Center, AMIST, College of Medicine, University of Ulsan, Seoul, Korea
| | - So-Yeon Yun
- Asan Medical Center, AMIST, College of Medicine, University of Ulsan, Seoul, Korea
| | - Bu-Kyu Lee
- Biomedical Engineering Research Center, Asan Medical Center, Asan Institute for Life Sciences, Seoul, Korea.
- Asan Medical Center, AMIST, College of Medicine, University of Ulsan, Seoul, Korea.
- Department of Oral and Maxillofacial Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
41
|
Li W, He H, Du M, Gao M, Sun Q, Wang Y, Lu H, Ou S, Xia C, Xu C, Zhao Q, Sun H. Quercetin as a promising intervention for rat osteoarthritis by decreasing M1-polarized macrophages via blocking the TRPV1-mediated P2X7/NLRP3 signaling pathway. Phytother Res 2024; 38:1990-2006. [PMID: 38372204 DOI: 10.1002/ptr.8158] [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: 09/19/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/20/2024]
Abstract
Osteoarthritis (OA) is characterized by an imbalance between M1 and M2 polarized synovial macrophages. Quercetin has shown protective effects against OA by altering M1/M2-polarized macrophages, but the underlying mechanisms remain unclear. In this study, rat chondrocytes were treated with 10 ng/mL of IL-1β. To create M1-polarized macrophages in vitro, rat bone marrow-derived macrophages (rBMDMs) were treated with 100 ng/mL LPS. To mimic OA conditions observed in vivo, a co-culture system of chondrocytes and macrophages was established. ATP release assays, immunofluorescence assays, Fluo-4 AM staining, Transwell assays, ELISA assays, and flow cytometry were performed. Male adult Sprague-Dawley (SD) rats were used to create an OA model. Histological analyses, including H&E, and safranin O-fast green staining were performed. Our data showed a quercetin-mediated suppression of calcium ion influx and ATP release, with concurrent downregulation of TRPV1 and P2X7 in the chondrocytes treated with IL-1β. Activation of TRPV1 abolished the quercetin-mediated effects on calcium ion influx and ATP release in chondrocytes treated with IL-1β. In the co-culture system, overexpression of P2X7 in macrophages attenuated the quercetin-mediated effects on M1 polarization, migration, and inflammation. Either P2X7 or NLRP3 knockdown attenuated IL-1β-induced M1/M2 polarization, migration, and inflammation. Moreover, overexpression of TRPV1 reduced the quercetin-mediated suppressive effects on OA by promoting M1/M2-polarized macrophages in vivo. Collectively, our data showed that quercetin-induced suppression of TRPV1 leads to a delay in OA progression by shifting the macrophage polarization from M1 to M2 subtypes via modulation of the P2X7/NLRP3 pathway.
Collapse
Affiliation(s)
- Wenjun Li
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Hebei He
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Min Du
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Mu Gao
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Qijie Sun
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Yeyang Wang
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Hanyu Lu
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Shuanji Ou
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Changliang Xia
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Changpeng Xu
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Qi Zhao
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Hongtao Sun
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
42
|
Kong H, Han JJ, Dmitrii G, Zhang XA. Phytochemicals against Osteoarthritis by Inhibiting Apoptosis. Molecules 2024; 29:1487. [PMID: 38611766 PMCID: PMC11013217 DOI: 10.3390/molecules29071487] [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: 02/27/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease that causes pathological changes in articular cartilage, synovial membrane, or subchondral bone. Conventional treatments for OA include surgical and non-surgical methods. Surgical treatment is suitable for patients in the terminal stage of OA. It is often the last choice because of the associated risks and high cost. Medication of OA mainly includes non-steroidal anti-inflammatory drugs, analgesics, hyaluronic acid, and cortico-steroid anti-inflammatory drugs. However, these drugs often have severe side effects and cannot meet the needs of patients. Therefore, safe and clinically appropriate long-term treatments for OA are urgently needed. Apoptosis is programmed cell death, which is a kind of physiologic cell suicide determined by heredity and conserved by evolution. Inhibition of apoptosis-related pathways has been found to prevent and treat a variety of diseases. Excessive apoptosis can destroy cartilage homeostasis and aggravate the pathological process of OA. Therefore, inhibition of apoptosis-related factors or signaling pathways has become an effective means to treat OA. Phytochemicals are active ingredients from plants, and it has been found that phytochemicals can play an important role in the prevention and treatment of OA by inhibiting apoptosis. We summarize preclinical and clinical studies of phytochemicals for the treatment of OA by inhibiting apoptosis. The results show that phytochemicals can treat OA by targeting apoptosis-related pathways. On the basis of improving some phytochemicals with low bioavailability, poor water solubility, and high toxicity by nanotechnology-based drug delivery systems, and at the same time undergoing strict clinical and pharmacological tests, phytochemicals can be used as a potential therapeutic drug for OA and may be applied in clinical settings.
Collapse
Affiliation(s)
- Hui Kong
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.K.); (J.-J.H.)
| | - Juan-Juan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.K.); (J.-J.H.)
| | - Gorbachev Dmitrii
- General Hygiene Department, Samara State Medical University, Samara 443000, Russia;
| | - Xin-an Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.K.); (J.-J.H.)
| |
Collapse
|
43
|
Li L, Fan Q, Zhao Y, Zhang Q, Qin G, Li C, Li W. Gentiopicroside ameliorates the lipopolysaccharide-induced inflammatory response and hypertrophy in chondrocytes. J Orthop Surg Res 2024; 19:198. [PMID: 38528538 DOI: 10.1186/s13018-024-04676-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/14/2024] [Indexed: 03/27/2024] Open
Abstract
PURPOSE This study aimed to evaluate the protective effects of gentiopicroside against lipopolysaccharide-induced chondrocyte inflammation. METHODS SW 1353 chondrosarcoma cells were stimulated with LPS (5 μg/ml) for 24 h and treated with different concentrations of gentiopicroside (GPS) for 24 h. The toxic effects of GPS on chondrocytes were determined using a CCK-8 assay and EdU staining. Western blotting, qPCR, and immunofluorescence analysis were used to examine the protective effect of GPS against the inflammatory response in chondrocytes induced by lipopolysaccharide (LPS). One-way ANOVA was used to compare the differences between the groups (significance level of 0.05). RESULTS The CCK-8 results showed that 10, 20 and 40 μM GPS had no significant toxic effects on chondrocytes; GPS effectively reduced the production of IL-1β and PGE2, reversed LPS-induced extracellular matrix degradation in cartilage by inhibiting the Stat3/Runx2 signaling pathway, and suppressed the hypertrophic transformation of SW 1353 chondrosarcoma cells. CONCLUSION Our study demonstrated that GPS significantly inhibited the LPS-induced inflammatory response and hypertrophic cellular degeneration in SW 1353 chondrosarcoma cells and is a valuable traditional Chinese medicine for the treatment of knee osteoarthritis.
Collapse
Affiliation(s)
- Longfei Li
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai, Shandong, China
| | - Qianqian Fan
- Department of Rehabilitation, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yixuan Zhao
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai, Shandong, China
| | - Qian Zhang
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai, Shandong, China
| | - Gaofeng Qin
- Department of Rehabilitation, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Department of Rehabilitation, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Wei Li
- Department of Rehabilitation, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
44
|
Yang SY, Hu Y, Zhao R, Zhou YN, Zhuang Y, Zhu Y, Ge XL, Lu TW, Lin KL, Xu YJ. Quercetin-loaded mesoporous nano-delivery system remodels osteoimmune microenvironment to regenerate alveolar bone in periodontitis via the miR-21a-5p/PDCD4/NF-κB pathway. J Nanobiotechnology 2024; 22:94. [PMID: 38449005 PMCID: PMC10918894 DOI: 10.1186/s12951-024-02352-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 02/20/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Impaired osteo-/angiogenesis, excessive inflammation, and imbalance of the osteoimmune homeostasis are involved in the pathogenesis of the alveolar bone defect caused by periodontitis. Unfortunately, there is still a lack of ideal therapeutic strategies for periodontitis that can regenerate the alveolar bone while remodeling the osteoimmune microenvironment. Quercetin, as a monomeric flavonoid, has multiple pharmacological activities, such as pro-regenerative, anti-inflammatory, and immunomodulatory effects. Despite its vast spectrum of pharmacological activities, quercetin's clinical application is limited due to its poor water solubility and low bioavailability. RESULTS In this study, we fabricated a quercetin-loaded mesoporous bioactive glass (Quercetin/MBG) nano-delivery system with the function of continuously releasing quercetin, which could better promote the bone regeneration and regulate the immune microenvironment in the alveolar bone defect with periodontitis compared to pure MBG treatment. In particular, this nano-delivery system effectively decreased injection frequency of quercetin while yielding favorable therapeutic results. In view of the above excellent therapeutic effects achieved by the sustained release of quercetin, we further investigated its therapeutic mechanisms. Our findings indicated that under the periodontitis microenvironment, the intervention of quercetin could restore the osteo-/angiogenic capacity of periodontal ligament stem cells (PDLSCs), induce immune regulation of macrophages and exert an osteoimmunomodulatory effect. Furthermore, we also found that the above osteoimmunomodulatory effects of quercetin via macrophages could be partially blocked by the overexpression of a key microRNA--miR-21a-5p, which worked through inhibiting the expression of PDCD4 and activating the NF-κB signaling pathway. CONCLUSION In summary, our study shows that quercetin-loaded mesoporous nano-delivery system has the potential to be a therapeutic approach for reconstructing alveolar bone defects in periodontitis. Furthermore, it also offers a new perspective for treating alveolar bone defects in periodontitis by inhibiting the expression of miR-21a-5p in macrophages and thereby creating a favorable osteoimmune microenvironment.
Collapse
Affiliation(s)
- Shi-Yuan Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yue Hu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Ran Zhao
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Ning Zhou
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yu Zhuang
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xiao-Li Ge
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting-Wei Lu
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai-Li Lin
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China.
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuan-Jin Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
- College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai Research Institute of Stomatology, Shanghai, China.
| |
Collapse
|
45
|
Mao H, Feng Y, Feng J, Yusufu Y, Sun M, Yang L, Jiang Q. Quercetin-3-O-β-D-glucuronide attenuates osteoarthritis by inhibiting cartilage extracellular matrix degradation and inflammation. J Orthop Translat 2024; 45:236-246. [PMID: 38601200 PMCID: PMC11004501 DOI: 10.1016/j.jot.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/21/2024] [Accepted: 01/31/2024] [Indexed: 04/12/2024] Open
Abstract
Objective Osteoarthritis (OA) is a chronic degenerative joint disease characterized by cartilage damage. In order to find a safer and more effective drug to treat OA, we investigated the role of quercetin-3-O-β-D-glucuronide (Q3GA) in OA. Methods We used qRT-PCR and western blots to detect the effects of Q3GA on extracellular matrix (ECM) and inflammation related genes and proteins in interleukin-1β (IL-1β) induced chondrocytes. We determined the effect of Q3GA on the NF-κB pathway using western blots and immunofluorescence. Moreover, the effect of Q3GA on the Nrf2 pathway was evaluated through molecular docking, western blots, and immunofluorescence experiments and further validated by transfection with Nrf2 siRNA. Subsequently, we established a rat model of OA and injected Q3GA into the joint cavity for treatment. After 5 weeks of Q3GA administration, samples were obtained for micro-computed tomography scanning and histopathological staining to determine the effects of Q3GA on OA rats. Results We found that Q3GA reduced the degradation of ECM and the expression of inflammatory related proteins and genes in primary chondrocytes of rats induced by IL-1β, as well as the expression of nitric oxide (NO) and reactive oxygen species (ROS). It inhibited the activation of the NF-κB pathway by increasing the expression of Nrf2 in the nucleus. In addition, Q3GA inhibited cartilage degradation in OA rats and promoted cartilage repair. Conclusion Q3GA attenuates OA by inhibiting ECM degradation and inflammation via the Nrf2/NF-κB axis. The translational potential of this article The results of our study demonstrate the promising potential of Q3GA as a candidate drug for the treatment of OA and reveal its key mechanisms.
Collapse
Affiliation(s)
- Haijun Mao
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
| | - Yanwei Feng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Juan Feng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yalikun Yusufu
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, China
| | - Minghui Sun
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, China
| | - Lei Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qing Jiang
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, China
| |
Collapse
|
46
|
Yuan Z, Jiang D, Yang M, Tao J, Hu X, Yang X, Zeng Y. Emerging Roles of Macrophage Polarization in Osteoarthritis: Mechanisms and Therapeutic Strategies. Orthop Surg 2024; 16:532-550. [PMID: 38296798 PMCID: PMC10925521 DOI: 10.1111/os.13993] [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: 10/11/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 02/02/2024] Open
Abstract
Osteoarthritis (OA) is the most common chronic degenerative joint disease in middle-aged and elderly people, characterized by joint pain and dysfunction. Macrophages are key players in OA pathology, and their activation state has been studied extensively. Various studies have suggested that macrophages might respond to stimuli in their microenvironment by changing their phenotypes to pro-inflammatory or anti-inflammatory phenotypes, which is called macrophage polarization. Macrophages accumulate and become polarized (M1 or M2) in many tissues, such as synovium, adipose tissue, bone marrow, and bone mesenchymal tissues in joints, while resident macrophages as well as other stromal cells, including fibroblasts, chondrocytes, and osteoblasts, form the joint and function as an integrated unit. In this study, we focus exclusively on synovial macrophages, adipose tissue macrophages, and osteoclasts, to investigate their roles in the development of OA. We review recent key findings related to macrophage polarization and OA, including pathogenesis, molecular pathways, and therapeutics. We summarize several signaling pathways in macrophage reprogramming related to OA, including NF-κB, MAPK, TGF-β, JAK/STAT, PI3K/Akt/mTOR, and NLRP3. Of note, despite the increasing availability of treatments for osteoarthritis, like intra-articular injections, surgery, and cellular therapy, the demand for more effective clinical therapies has remained steady. Therefore, we also describe the current prospective therapeutic methods that deem macrophage polarization to be a therapeutic target, including physical stimulus, chemical compounds, and biological molecules, to enhance cartilage repair and alleviate the progression of OA.
Collapse
Affiliation(s)
- Zimu Yuan
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Decheng Jiang
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Mengzhu Yang
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Jie Tao
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Xin Hu
- Orthopedic Research Institute, Department of OrthopedicsWest China Hospital, Sichuan UniversityChengduChina
| | - Xiao Yang
- National Engineering Research Center for BiomaterialsSichuan UniversityChengduChina
| | - Yi Zeng
- Orthopedic Research Institute, Department of OrthopedicsWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
47
|
Hu Y, Fu Z, Yang S, Zhou Y, Zhu H, Zhu Y, Zhou J, Lin K, Xu Y. A multifunctional quercetin/polycaprolactone electrospun fibrous membrane for periodontal bone regeneration. Mater Today Bio 2024; 24:100906. [PMID: 38226016 PMCID: PMC10788537 DOI: 10.1016/j.mtbio.2023.100906] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 01/17/2024] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- Yue Hu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zeyu Fu
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, China
| | - Shiyuan Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yuning Zhou
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Huimin Zhu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yan Zhu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jia Zhou
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Kaili Lin
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yuanjin Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
48
|
Li G, Ren Y, Li E, Deng K, Qu C, Zhang J, Zhang L, Wang X, Lian J, Zhou H, Wang Z, Shen T, Li X, Jiang Z. Quercetin inhibits mesothelial-mesenchymal transition and alleviates postoperative peritoneal adhesions by blocking the TGF-β1/PI3K/AKT pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117242. [PMID: 37777024 DOI: 10.1016/j.jep.2023.117242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Carthami flos is a dried flower of the Asteraceae plant Carthamus tinctorius (L.). Danhong injection, composed of Carthami flos and Danshen can prevent the formation of postoperative peritoneal adhesions. Quercetin (QUE), an active compound of Carthami flos, has also been proved to prevent postoperative abdominal and uterine cavity adhesions. However, whether QUE is the key component in Carthami flos and the mechanism in preventing postoperative peritoneal adhesions has not been studied. AIM OF THE STUDY To predict whether QUE is the key molecule in Carthami flos and explore the effect and mechanism of QUE in preventing postoperative peritoneal adhesions. MATERIALS AND METHODS Drug composition and target analysis was used to predict the key component in Carthami flos. The method of cecum-sidewall abrasion was used to establish adhesion models, and the antiadhesion effect of QUE was evaluated with the adhesion scoring system. Network pharmacology was used to predict the targets and potential mechanism of QUE in preventing adhesion. The mechanism was further verified by immunofluorescence, Western blot, wound healing experiment, and molecular docking. RESULTS Quercetin was predicted to be the key to preventing postoperative peritoneal adhesions in Carthami flos. Animal experiments revealed that QUE effectively ameliorated adhesions and reduced the expression of mesothelial-mesenchymal transition (MMT) related markers and TGF-β1. Moreover, the TGF-β1/PI3K/AKT pathway was predicted via protein-protein interaction and Kyoto encyclopedia of Genes and Genomes enrichment analysis to play a crucial part in preventing adhesion by QUE. Furthermore, in vitro experiments and molecular docking demonstrated that QUE could block the TGF-β1/PI3K/AKT pathway through forming a stable combination with TβR-II, thereby inhibiting MMT and ameliorating peritoneal adhesion. CONCLUSIONS QUE can not only reduce postoperative TGF-β1 but also block the TGF-β1/PI3K/AKT pathway to inhibit MMT of mesothelial cells, and finally alleviate postoperative peritoneal adhesions. These findings may provide insights towards development of a safe and effective anti-adhesive drug for prevention of postoperative peritoneal adhesions.
Collapse
Affiliation(s)
- Gan Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yiwei Ren
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Enmeng Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Kai Deng
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Chao Qu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Junxiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Li Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xingjie Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jie Lian
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Huayou Zhou
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China; Department of General Surgery, Hanzhong Central Hospital, Hanzhong, 723000, Shaanxi, China
| | - Zijun Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Tianli Shen
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China; Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Zhengdong Jiang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
49
|
Deng TT, Ding WY, Lu XX, Zhang QH, Du JX, Wang LJ, Yang MN, Yin Y, Liu FJ. Pharmacological and mechanistic aspects of quercetin in osteoporosis. Front Pharmacol 2024; 15:1338951. [PMID: 38333006 PMCID: PMC10851760 DOI: 10.3389/fphar.2024.1338951] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Osteoporosis (OP) is a bone disease associated with increasing age. Currently, the most common medications used to treat OP are anabolic agents, anti-resorptive agents, and medications with other mechanisms of action. However, many of these medications have unfavorable adverse effects or are not intended for long-term use, potentially exerting a severe negative impact on a patient's life and career and placing a heavy burden on families and society. There is an urgent need to find new drugs that can replace these and have fewer adverse effects. Quercetin (Que) is a common flavonol in nature. Numerous studies have examined the therapeutic applications of Que. However, a comprehensive review of the anti-osteoporotic effects of Que has not yet been conducted. This review aimed to describe the recent studies on the anti-osteoporotic effects of Que, including its biological, pharmacological, pharmacokinetic, and toxicological properties. The outcomes demonstrated that Que could enhance OP by increasing osteoblast differentiation and activity and reducing osteoclast differentiation and activity via the pathways of Wnt/β-catenin, BMP/SMAD/RUNX2, OPG/RANKL/RANK, ERK/JNK, oxidative stress, apoptosis, and transcription factors. Thus, Que is a promising novel drug for the treatment of OP.
Collapse
Affiliation(s)
- Ting-Ting Deng
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Yu Ding
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, China
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xi-Xue Lu
- Bone Biomechanics Engineering Laboratory of Shandong Province, Shandong Medicinal Biotechnology Center, School of Biomedical Sciences, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qing-Hao Zhang
- Bone Biomechanics Engineering Laboratory of Shandong Province, Shandong Medicinal Biotechnology Center, School of Biomedical Sciences, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jin-Xin Du
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Li-Juan Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Bone Biomechanics Engineering Laboratory of Shandong Province, Shandong Medicinal Biotechnology Center, School of Biomedical Sciences, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Mei-Na Yang
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Biomedical Sciences College, Shandong First Medical University, Jinan, China
| | - Ying Yin
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fan-Jie Liu
- Bone Biomechanics Engineering Laboratory of Shandong Province, Shandong Medicinal Biotechnology Center, School of Biomedical Sciences, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
50
|
Chen K, Tao H, Zhu P, Chu M, Li X, Shi Y, Zhang L, Xu Y, Lv S, Huang L, Huang W, Geng D. ADAM8 silencing suppresses the migration and invasion of fibroblast-like synoviocytes via FSCN1/MAPK cascade in osteoarthritis. Arthritis Res Ther 2024; 26:20. [PMID: 38218854 PMCID: PMC10787439 DOI: 10.1186/s13075-023-03238-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/13/2023] [Indexed: 01/15/2024] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a degenerative joint disease that affects elderly populations worldwide, causing pain and disability. Alteration of the fibroblast-like synoviocytes (FLSs) phenotype leads to an imbalance in the synovial inflammatory microenvironment, which accelerates the progression of OA. Despite this knowledge, the specific molecular mechanisms of the synovium that affect OA are still unclear. METHODS Both in vitro and in vivo experiments were undertaken to explore the role of ADAM8 playing in the synovial inflammatory of OA. A small interfering RNA (siRNA) was targeting ADAM8 to intervene. High-throughput sequencing was also used. RESULTS Our sequencing analysis revealed significant upregulation of the MAPK signaling cascade and ADAM8 gene expression in IL-1β-induced FLSs. The in vitro results demonstrated that ADAM8 blockade inhibited the invasion and migration of IL-1β-induced FLSs, while also suppressing the expression of related matrix metallomatrix proteinases (MMPs). Furthermore, our study revealed that inhibiting ADAM8 weakened the inflammatory protein secretion and MAPK signaling networks in FLSs. Mechanically, it revealed that inhibiting ADAM8 had a significant effect on the expression of migration-related signaling proteins, specifically FSCN1. When siADAM8 was combined with BDP-13176, a FSCN1 inhibitor, the migration and invasion of FLSs was further inhibited. These results suggest that FSCN1 is a crucial downstream factor of ADAM8 in regulating the biological phenotypes of FLSs. The in vivo experiments demonstrated that ADAM8 inhibition effectively reduced synoviocytes inflammation and alleviated the progression of OA in rats. CONCLUSIONS ADAM8 could be a promising therapeutic target for treating OA by targeting synovial inflammation.
Collapse
Affiliation(s)
- Kai Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, Jiangsu, China
- Department of Orthopedics, Hai'an People's Hospital, Zhongba Road 17, Hai'an, Jiangsu, China
| | - Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, Jiangsu, China
| | - Pengfei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, Jiangsu, China
| | - Miao Chu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, Jiangsu, China
- Department of Orthopedics, Yixing Peoples's Hospital, Xincheng Road 1588, Yixing, Jiangsu, China
| | - Xueyan Li
- Anesthesiology department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Guangjj Road 242, Suzhou, Jiangsu, China
| | - Yi Shi
- Anesthesiology department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Guangjj Road 242, Suzhou, Jiangsu, China
| | - Liyuan Zhang
- Anesthesiology department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Guangjj Road 242, Suzhou, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, Jiangsu, China
| | - Shujun Lv
- Department of Orthopedics, Hai'an People's Hospital, Zhongba Road 17, Hai'an, Jiangsu, China.
| | - Lixin Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, Jiangsu, China.
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road 17, Hefei, An'hui, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, Jiangsu, China.
| |
Collapse
|