1
|
Skalny AV, Menshikova IV, Korobeinikova TV, Morozova GD, Guo X, Zhang F, Tinkov AA. Circulating Essential Trace Element and Mineral Levels in Female Patients with Knee or Concomitant Knee and Hip Osteoarthritis. Biol Trace Elem Res 2025:10.1007/s12011-025-04696-w. [PMID: 40490590 DOI: 10.1007/s12011-025-04696-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Accepted: 06/04/2025] [Indexed: 06/11/2025]
Abstract
The objective of the present study was to evaluate serum trace element and mineral levels in female patients with knee osteoarthritis (kOA) and concomitant knee and hip osteoarthritis (khOA). The study enrolled 61 healthy women without OA or any other pathology of joints (OA-free group), 180 subjects with bilateral kOA, and 99 patients with khOA. Age, anthropometric parameters, and the prevalence of concomitant diseases were registered. Serum trace element and mineral levels were assessed using inductively-coupled plasma mass-spectrometry. The obtained data demonstrate that patients with kOA and especially khOA are characterized by higher age, body mass index, and the prevalence of hypertension and certain other comorbidities. Serum analysis demonstrated that Zn and Fe levels in kOA and khOA patients were lower than those in OA-free patients by 7% and 9%, and 14% and 18%, respectively. In turn, circulating Se concentrations in patients with kOA and khOA were 9% and 7% lower compared to OA-free group values. Serum Cu and Mo levels in khOA patients exceeded the respective values in kOA subjects. Factorial analysis demonstrated that OA case status had a significant impact on serum Ca, Cr, Fe, Mn, Se, V, and Zn. At the same time, multiple regression analysis revealed a significant association with OA only for serum Zn, Se, V after adjustment for age, BMI, and the prevalence of concomitant diseases. Therefore, the obtained data indicate that involvement of multiple joints in OA patients is associated with more severe alterations in trace element levels.
Collapse
Affiliation(s)
- Anatoly V Skalny
- Center of Bioelementology and Human Ecology, and the Department of Hospital Therapy No. 1, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia.
- Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia.
| | - Irina V Menshikova
- Center of Bioelementology and Human Ecology, and the Department of Hospital Therapy No. 1, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia
| | - Tatiana V Korobeinikova
- Center of Bioelementology and Human Ecology, and the Department of Hospital Therapy No. 1, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia
- Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Galina D Morozova
- Center of Bioelementology and Human Ecology, and the Department of Hospital Therapy No. 1, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia
| | - Xiong Guo
- Key Laboratory of Trace Elements and Endemic Diseases, Health Science Center, School of Public Health, National Health and Family Planning Commission, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Health Science Center, School of Public Health, National Health and Family Planning Commission, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Alexey A Tinkov
- Center of Bioelementology and Human Ecology, and the Department of Hospital Therapy No. 1, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia
- Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150000, Russia
| |
Collapse
|
2
|
Khanal V, Carroll M, Carter J, Zhong Y, Chikkamagaluru S, Sato A, Allen R, Wankhade U, Dole N. Lipocalin-2 Regulates Osteocyte Ferroptosis and Osteocyte-Osteoblast Crosstalk via Wnt Signaling to Control Bone Formation. RESEARCH SQUARE 2025:rs.3.rs-6430607. [PMID: 40343339 PMCID: PMC12060985 DOI: 10.21203/rs.3.rs-6430607/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Osteoporosis is a multifactorial disease, and emerging evidence suggests that iron overload contributes to its progression. Here, we identify Lipocalin-2 (LCN2), a cytokine secreted by bone cells with endocrine effects on other tissues, as a local regulator of osteocyte iron metabolism and a mediator of skeletal deterioration. Our findings reveal that LCN2 promotes iron accumulation, mitochondrial dysfunction, and ferroptosis in osteocytes in a process dependent on LCN2 receptor SLC22A17. Genetic ablation of Lcn2 (Dmp1-Cre; Lcn2 fl/fl ) in osteocytes mitigates their ferroptotic vulnerability by preserving mitochondrial integrity and limiting iron overload. Remarkably, LCN2 deletion enhances osteocyte dendricity and lacunocanalicular network, supporting their function in bone remodeling. Mechanistically, we demonstrate that Lcn2 ablation in osteocytes decreases DKK1 and SOST expression in bone, leading to increased Wnt/β-catenin signaling and osteoblast-driven bone formation. Using in vitro and in vivo approaches, we establish the LCN2-SLC22A17 axis as a key pathway linking iron homeostasis, osteocyte dysfunction, and skeletal remodeling. These findings provide insight into a previously unrecognized mechanism underlying iron-driven bone loss and suggest that targeting LCN2 could offer therapeutic potential for osteoporosis.
Collapse
Affiliation(s)
| | | | | | - Ying Zhong
- University of Arkansas for Medical Sciences
| | | | - Amy Sato
- University of Arkansas for Medical Sciences
| | - Ryan Allen
- University of Arkansas for Medical Sciences
| | | | - Neha Dole
- University of Arkansas for Medical Sciences
| |
Collapse
|
3
|
Ioannidis K, Dimopoulos A, Decoene I, Guilliams M, Svitina H, Storozhuk L, de Oliveira‐Silva R, Basov S, Thanh NTK, Mourdikoudis S, Van Bael MJ, Smeets B, Sakellariou D, Papantoniou I. 4D Biofabrication of Magnetically Augmented Callus Assembloid Implants Enables Rapid Endochondral Ossification via Activation of Mechanosensitive Pathways. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413680. [PMID: 39998420 PMCID: PMC12005758 DOI: 10.1002/advs.202413680] [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: 10/25/2024] [Revised: 01/31/2025] [Indexed: 02/26/2025]
Abstract
The use of magnetic-driven strategies for non-contact manipulation of engineered living modules opens up new possibilities for tissue engineering. The integration of magnetic nanoparticles (MNPs) with cartilaginous microtissues enables model-driven 4D bottom-up biofabrication of remotely actuated assembloids, providing unique properties to mechanoresponsive tissues, particularly skeletal constructs. However, for clinical use, the long-term effects of magnetic stimulation on phenotype and in vivo functionality need further exploration. Magnetic-driven biofabrication includes both rapid processes, such as guided microtissue assembly, and slower biological processes, like extracellular matrix secretion. This work explores the interplay between magnetic fields and MNP-loaded cartilaginous microtissues through mathematical modeling and experimental approaches, investigating long-term stimulation effects on ECM maturation and chondrogenic hypertrophy. Transcriptomic analysis reveal that magnetic stimulation activated mechanosensitive pathways and catabolic processes, driving accelerated cartilage-to-bone transitions via endochondral ossification, outcomes not observed in non-stimulated controls. This study paves the way for pre-programmed, remotely actuated skeletal assembloids with superior bone-forming capacity for regenerating challenging bone fractures.
Collapse
Affiliation(s)
- Konstantinos Ioannidis
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Andreas Dimopoulos
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Isaak Decoene
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Maya Guilliams
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
- MeBioS division, Biosystems DepartmentKU LeuvenKasteelpark, Arenberg 30Leuven3001Belgium
| | - Hanna Svitina
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Liudmyla Storozhuk
- Healthcare Biomagnetics and Nanomaterials Laboratories, Department of Medical Physics and Biomedical EngineeringUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- London Centre for NanotechnologyUniversity College London17‐19 Gordon StreetLondonWC1H 0AHUK
| | - Rodrigo de Oliveira‐Silva
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular SystemsKU LeuvenCelestijnenlaan 200F, PB 2454Leuven3001Belgium
| | - Sergey Basov
- Quantum Solid State Physics, Department of Physics and AstronomyKU LeuvenCelestijnenlaan 200DLeuven3001Belgium
| | - Nguyen Thi Kim Thanh
- Healthcare Biomagnetics and Nanomaterials Laboratories, Department of Medical Physics and Biomedical EngineeringUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- Biophysics Group, Department of Physics and AstronomyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Stefanos Mourdikoudis
- CINBIO, Department of Physical Chemistry, Campus Universitario, Lagoas MarcosendeUniversidade de VigoVigo36310Spain
| | - Margriet J. Van Bael
- Quantum Solid State Physics, Department of Physics and AstronomyKU LeuvenCelestijnenlaan 200DLeuven3001Belgium
| | - Bart Smeets
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
- MeBioS division, Biosystems DepartmentKU LeuvenKasteelpark, Arenberg 30Leuven3001Belgium
| | - Dimitrios Sakellariou
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular SystemsKU LeuvenCelestijnenlaan 200F, PB 2454Leuven3001Belgium
| | - Ioannis Papantoniou
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| |
Collapse
|
4
|
Yang RS, Chan DC, Chung YP, Liu SH. Chronic Kidney Disease and Osteoarthritis: Current Understanding and Future Research Directions. Int J Mol Sci 2025; 26:1567. [PMID: 40004032 PMCID: PMC11854965 DOI: 10.3390/ijms26041567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/02/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Chronic kidney disease (CKD) is a significant public health concern. Osteoarthritis (OA), a common form of arthritis, has been shown to have a dramatically increased prevalence, particularly among individuals aged 40-50 and older, in the presence of CKD. Furthermore, CKD may exacerbate the progression and impact of OA. A survey study revealed that 53.9% of CKD patients undergoing long-term hemodialysis were diagnosed with OA. These findings underscore the potential association between CKD and OA. Uremic toxins, such as indoxyl sulfate, p-cresyl sulfate, transforming growth factor-β, and advanced glycation end-products, are regarded as potential risk factors in various CKD-related conditions, affecting bone and joint metabolism. However, whether these factors serve as a bridging mechanism between CKD and OA comorbidities, as well as their detailed roles in this context, remains unclear. Addressing the progression of OA in CKD patients and identifying effective treatment and prevention strategies is an urgent challenge that warrants immediate attention. This review focuses on describing and discussing the molecular pathological mechanisms underlying CKD-associated OA and the possible therapeutic strategies.
Collapse
Affiliation(s)
- Rong-Sen Yang
- Department of Orthopedics, College of Medicine and Hospital, National Taiwan University, Taipei 100, Taiwan;
| | - Ding-Cheng Chan
- Department of Geriatrics and Gerontology, College of Medicine and Hospital, National Taiwan University, Taipei 100, Taiwan;
| | - Yao-Pang Chung
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 406, Taiwan
- Department of Pediatrics, College of Medicine and Hospital, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
5
|
Prasadam I, Schrobback K, Kranz-Rudolph B, Fischer N, Sonar Y, Sun AR, Secondes E, Klein T, Crawford R, Subramaniam VN, Rishi G. Effects of iron overload in human joint tissue explant cultures and animal models. J Mol Med (Berl) 2025; 103:73-86. [PMID: 39531048 DOI: 10.1007/s00109-024-02495-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease affecting over 530 million individuals worldwide. Recent studies suggest a potential link between iron overload, a condition characterised by the excessive accumulation of iron in the body, and the onset of OA. Iron is essential for various biological processes, and any disruption in its homeostasis can trigger significant health effects, including OA. This study aimed to elucidate the effects of excess iron on joint tissue and the underlying mechanisms associated with excess iron and OA development. Human articular cartilage (n = 6) and synovium (n = 4) were collected from patients who underwent total knee arthroplasty. Cartilage and synovium explants were incubated with a gradually increasing concentration of ferric ammonium citrate for 3 days respectively. The effects of iron homeostasis in tissue explants were analysed using a Laser Ablation Inductively Coupled Plasma Mass Spectrometry (LA-ICP-MS). To further study the effects of iron excess on OA initiation and development, male 3-week-old Hfe-/- and 5-week-old Tfr2-/- mice, animal models of hereditary haemochromatosis were established. Littermate wild-type mice were fed a high-iron diet to induce dietary overload. All animals were sacrificed at 8 weeks of age, and knee joints were harvested for histological analysis. The LA-ICP-MS analysis unveiled changes in the elemental composition related to iron metabolism, which included alterations in FTH1, FPN1, and HAMP within iron(III)-treated cartilage explants. While chondrocyte viability remained stable under different iron concentrations, ex vivo treatment with a high concentration of Fe3+ increased the catabolic gene expression of MMP13. Similar alterations were observed in the synovium, with added increases in GAG content and inflammation markers. In vivo studies further supported the role of iron overload in OA development as evidenced by spontaneous OA symptoms, proteoglycan loss, increased Mankin scores, synovial thickening, and enhanced immunohistochemical expression of MMP13, ADAMTS5, and P21 in Hfe-/-, Tfr2-/-, and diet-induced iron overload mouse models. Our findings elucidate the specific pathways through which excess iron accelerates OA progression and highlights potential targets for therapeutic intervention aimed at modulating iron levels to mitigate OA symptoms. KEY MESSAGES: Iron overload alters joint iron metabolism, increasing OA markers in cartilage and synovium. High iron levels in mice accelerate OA, highlighting genetic and dietary impacts. Excess iron prompts chondrocyte iron storage response, signalling potential OA pathways. Iron dysregulation linked to increased cartilage degradation and synovial inflammation. Our findings support targeted therapies for OA based on iron modulation strategies.
Collapse
Affiliation(s)
- Indira Prasadam
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia.
| | - Karsten Schrobback
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Bastian Kranz-Rudolph
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Nadine Fischer
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Yogita Sonar
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Antonia RuJia Sun
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Eriza Secondes
- Hepatogenomics Research Group, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, 4059, Australia
| | - Travis Klein
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Ross Crawford
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
- Orthopedic Department, The Prince Charles Hospital, Brisbane, QLD, 4032, Australia
| | - V Nathan Subramaniam
- Hepatogenomics Research Group, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Qld, 4059, Australia
| | - Gautam Rishi
- Hepatogenomics Research Group, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Qld, 4059, Australia
| |
Collapse
|
6
|
Sun W, Sun J, Hu W, Luo C, Lu Z, He F, Zhao H, Zeng X, Cao D, Li J, Zhang C, Xia J. Sulforaphane inhibits multiple myeloma cell-induced osteoclast differentiation and macrophage proliferation by elevating ferroportin1. Cancer Chemother Pharmacol 2024; 95:3. [PMID: 39661165 DOI: 10.1007/s00280-024-04736-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
PURPOSE Osteolysis is a common complication in patients with multiple myeloma (MM). Our previous studies have demonstrated that MM cells can promote osteoclast differentiation of macrophages. In this study, we explored the effect of sulforaphane (SFN), a natural NRF2 activator found in broccoli, on MM cell-induced osteoclast differentiation. METHODS Conditional medium (CM) derived from MM cells was used to induce osteoclast differentiation, and TRAP staining was performed to examine osteoclast. Gene expression was detected by western blotting or real-time PCR. Cell counting and EdU staining were performed to test macrophage proliferation. RESULTS We showed that the CM of MM cells downregulated the expression of ferroportin1 (Fpn1), the only known iron exporter in vertebrate cells, thereby increasing cellular iron levels in murine macrophage cells RAW264.7. Deferoxamine (DFO), an iron chelator, effectively blocked MM cell CM-induced osteoclast differentiation and macrophage proliferation, suggesting that iron overload played a key role in these cellular events. Subsequent mechanistic investigations revealed that MM cell CM induced osteoclast differentiation and macrophage proliferation by activating the JNK/AP-1/NFATC1 pathway and PI3K/AKT pathway. SFN was found to increase Fpn1 expression, leading to decreased cellular iron levels in RAW264.7 cells activated by MM cell CM. Importantly, the osteoclast differentiation and macrophage proliferation induced by MM cell CM were significantly inhibited by SFN. CONCLUSION Altogether, our findings indicated that SFN inhibits MM cell-induced osteoclast differentiation and macrophage proliferation by elevating FPN1 levels. SFN could be a promising therapeutic strategy for MM-associated osteolysis.
Collapse
Affiliation(s)
- Weichu Sun
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingqi Sun
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wei Hu
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Cong Luo
- Department of Hematology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhongwei Lu
- Department of Hematology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fen He
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hongyan Zhao
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xi Zeng
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Deliang Cao
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Junjun Li
- Department of Hematology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chang Zhang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Jiliang Xia
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
7
|
Huo G, Lin Y, Liu L, He Y, Qu Y, Liu Y, Zhu R, Wang B, Gong Q, Han Z, Yin H. Decoding ferroptosis: transforming orthopedic disease management. Front Pharmacol 2024; 15:1509172. [PMID: 39712490 PMCID: PMC11659002 DOI: 10.3389/fphar.2024.1509172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
As a mechanism of cell death, ferroptosis has gained popularity since 2012. The process is distinguished by iron toxicity and phospholipid accumulation, in contrast to autophagy, apoptosis, and other cell death mechanisms. It is implicated in the advancement of multiple diseases across the body. Researchers currently know that osteosarcoma, osteoporosis, and other orthopedic disorders are caused by NRF2, GPX4, and other ferroptosis star proteins. The effective relief of osteoarthritis symptoms from deterioration has been confirmed by clinical treatment with multiple ferroptosis inhibitors. At the same time, it should be reminded that the mechanisms involved in ferroptosis that regulate orthopedic diseases are not currently understood. In this manuscript, we present the discovery process of ferroptosis, the mechanisms involved in ferroptosis, and the role of ferroptosis in a variety of orthopedic diseases. We expect that this manuscript can provide a new perspective on clinical diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Guanlin Huo
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lusheng Liu
- Department of Acupuncture and Moxibustion, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqi He
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Yi Qu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yang Liu
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Renhe Zhu
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Bo Wang
- Department of Orthopaedics, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Qing Gong
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Zhongyu Han
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongbing Yin
- Orthopedic Center, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
8
|
Altahla R, Alshorman J, Tao X. Ferroptosis plays a role in osteoarthritis. ALL LIFE 2024; 17. [DOI: 10.1080/26895293.2024.2391292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/28/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Ruba Altahla
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jamal Alshorman
- Department of Orthopedics, The Second Affiliated Hospital, Hubei University of Science and Technology, Xiang Ning, People’s Republic of China
| | - Xu Tao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
9
|
Liu F. Iron overload is positively associated with the incidence of osteoarthritis: A NHANES cross-sectional study. Medicine (Baltimore) 2024; 103:e40089. [PMID: 39470535 PMCID: PMC11520987 DOI: 10.1097/md.0000000000040089] [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: 07/28/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
With the aging of the global population and the increase in the number of people with conditions such as obesity, the incidence of osteoarthritis (OA) is increasing annually. Clinical studies have shown that excessive accumulation of iron in joints is associated with age-related OA. However, there have been no reports on the relationship between iron metabolism and osteoarthritis. A STROBE-compliant cross-sectional observational study, was carried out and analyzed from the National Health and Nutrition Examination Survey from 2001 to 2020, including data on serum iron, transferrin saturation, serum ferritin, total iron-binding capacity, and transferrin receptors, as well as data on osteoarthritis. This cross-sectional study was conducted to explore the relationship between serum iron levels, osteoarthritis, and related metabolic factors. By adjusting the model and using quantile logistic regression models, the interaction between human body iron content and the aforementioned variables was analyzed. A total of 56,323 participants over 5 cycles were assessed for iron levels. After adjusting the model for age, sex, race, education level, marital status, total energy intake, physical activity, drinking, BMI, smoking, hypertension, and diabetes, we found that in different quantile regression results, serum iron was associated with OA, Q4: OR = 1.231 (95%CI: 1.009-1.501, P < .05). Ferritin is associated with OA, Q2: OR = 1.309 (95%CI: 1.012-1.692, P < .05); Q3: OR = 1.424 (95%CI: 1.129-1.797, P < .01); Q4: OR = 1.280 (95%CI: 1.013-1.616, P < .05). This cross-sectional study found that serum iron and transferrin saturation levels were positively correlated with OA incidence, suggesting that iron overload is a risk factor for OA. Large-sample prospective cohort studies are needed to confirm the correlation between iron overload and OA.
Collapse
Affiliation(s)
- Fei Liu
- Department of Orthopedics, People’s Hospital of Bazhou, Xinjiang, Korla, China
| |
Collapse
|
10
|
Kim H, Ko DS. Effects of Iron, Copper, Zinc, and Magnesium on Chronic Widespread Pain: A Two-Sample Mendelian Randomization. J Clin Med 2024; 13:5908. [PMID: 39407968 PMCID: PMC11478258 DOI: 10.3390/jcm13195908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Chronic widespread pain (CWP) affects approximately 10% of the adult population globally, causing significant physical and psychological distress. Micronutrients, such as iron, copper, zinc, and magnesium, are essential in various physiological functions, and their imbalances may impact pain perception and chronic pain conditions. Methods: This study used Mendelian randomization (MR) to investigate the causal relationships between micronutrient levels and CWP. Data were obtained from genome-wide association studies (GWASs) for iron, copper, zinc, and magnesium, and CWP data were sourced from large-scale GWASs with 461,857 European participants. Genetic variants were used as instrumental variables to infer causal relationships, minimizing confounding factors. Results: MR analysis revealed a significant association between higher iron levels and an increased risk of CWP (IVW, OR 1.01, 95% CI: 1.00-1.01, p = 0.029). This finding was supported by the weighted median and MR-Egger methods. No significant associations were found for copper, zinc, and magnesium levels. Conclusions: These results suggest that iron levels may influence pain perception and chronic pain conditions. Balanced iron levels are crucial for managing chronic pain. Regular monitoring and personalized treatment plans could benefit individuals with CWP. Further research is needed to understand the mechanisms linking micronutrient levels to chronic pain and to develop targeted therapeutic interventions.
Collapse
Affiliation(s)
- Hyunjik Kim
- Department of General Surgery, Breast Cancer Center, Gachon University Gil Medical Center, Incheon 21565, Republic of Korea
| | - Dai Sik Ko
- Division of Vascular Surgery, Department of General Surgery, Gachon University College of Medicine, Gil Medical Center, Incheon 21565, Republic of Korea
| |
Collapse
|
11
|
Zhang Y, Hu K, Shang Z, Yang X, Cao L. Ferroptosis: Regulatory mechanisms and potential targets for bone metabolism: A review. Medicine (Baltimore) 2024; 103:e39158. [PMID: 39331895 PMCID: PMC11441915 DOI: 10.1097/md.0000000000039158] [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: 11/15/2023] [Accepted: 07/10/2024] [Indexed: 09/29/2024] Open
Abstract
Bone homeostasis is a homeostasis process constructed by osteoblast bone formation and osteoclast bone resorption. Bone homeostasis imbalance and dysfunction are the basis for the development of various orthopedic diseases such as osteoporosis, osteoarthritis, and steroid-induced avascular necrosis of femoral head. Previous studies have demonstrated that ferroptosis can induce lipid peroxidation through the generation of reactive oxygen species, activate a number of signaling pathways, and participate in the regulation of osteoblast bone formation and osteoclast bone resorption, resulting in bone homeostasis imbalance, which is an important factor in the pathogenesis of many orthopedic diseases, but the mechanism of ferroptosis is still unknown. In recent years, it has been found that, in addition to iron metabolism and intracellular antioxidant system imbalance, organelle dysfunction is also a key factor affecting ferroptosis. This paper takes this as the starting point, reviews the latest literature reports at home and abroad, elaborates the pathogenesis and regulatory pathways of ferroptosis and the relationship between ferroptosis and various organelles, and summarizes the mechanism by which ferroptosis mediates bone homeostasis imbalance, with the aim of providing new directions for the research related to ferroptosis and new ideas for the prevention and treatment of bone and joint diseases.
Collapse
Affiliation(s)
- Yongjie Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Kangyi Hu
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhengya Shang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaorui Yang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linzhong Cao
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
- The Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
12
|
Cao S, Wei Y, Yue Y, Chen Y, Qian J, Wang D, Xiong A, Liu P, Zeng H. Rosiglitazone retards the progression of iron overload-induced osteoarthritis by impeding chondrocyte ferroptosis. iScience 2024; 27:110526. [PMID: 39224514 PMCID: PMC11366908 DOI: 10.1016/j.isci.2024.110526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/03/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Ferroptosis is implicated in several diseases, including iron overload-induced osteoarthritis (IOOA), which is marked by oxidative stress, iron imbalance, and lipid peroxidation. Given rosiglitazone's (RSG) ability to inhibit lipid peroxidation and ferroptosis, this study aims to assess its therapeutic potential for treating IOOA. Our in vitro results show that RSG targets acyl-CoA synthetase long-chain family member 4 to mitigate impairments induced by interleukin-1 beta and ferric ammonium citrate, including cell apoptosis, senescence, inflammatory responses, extracellular matrix degradation, and ferroptosis. RSG reduced intracellular iron content, alleviated oxidative stress and lipid peroxidation, mitigated damage to membrane-bound organelles, and enhanced glucose transport. Additionally, pre-treatment with RSG imparted anti-ferroptotic properties to chondrocytes. In vivo, RSG alleviated cartilage degradation, inflammatory responses, and ferroptosis in mice with IOOA. In conclusion, RSG exhibits chondroprotective and anti-ferroptotic effects by suppressing lipid peroxidation and restoring iron homeostasis, highlighting its potential for treating IOOA.
Collapse
Affiliation(s)
- Siyang Cao
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Yihao Wei
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Yaohang Yue
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Yingqi Chen
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Junyu Qian
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Deli Wang
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Ao Xiong
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Peng Liu
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Hui Zeng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| |
Collapse
|
13
|
Chen BY, Pathak JL, Lin HY, Guo WQ, Chen WJ, Luo G, Wang LJ, Sun XF, Ding Y, Li J, Diekwisch TGH, Liu C. Inflammation Triggers Chondrocyte Ferroptosis in TMJOA via HIF-1α/TFRC. J Dent Res 2024; 103:712-722. [PMID: 38766865 DOI: 10.1177/00220345241242389] [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: 05/22/2024] Open
Abstract
Inflammation and loss of articular cartilage are considered the major cause of temporomandibular joint osteoarthritis (TMJOA), a painful condition of the temporomandibular joint (TMJ). To determine the cause of TMJ osteoarthritis in these patients, synovial fluid of TMJOA patients was compared prior to and after hyaluronic lavage, revealing substantially elevated levels of interleukin (IL) 1β, reactive oxidative stress (ROS), and an overload of Fe3+ and Fe2+ prior to lavage, indicative of ferroptosis as a mode of chondrocyte cell death. To ask whether prolonged inflammatory conditions resulted in ferroptosis-like transformation in vitro, we subjected TMJ chondrocytes to IL-1β treatment, resulting in a shift in messenger RNA sequencing gene ontologies related to iron homeostasis and oxidative stress-related cell death. Exposure to rat unilateral anterior crossbite conditions resulted in reduced COL2A1 expression, fewer chondrocytes, glutathione peroxidase 4 (GPX4) downregulation, and 4-hydroxynonenal (4-HNE) upregulation, an effect that was reversed after intra-articular injections of the ferroptosis inhibitor ferrostatin 1 (Fer-1). Our study demonstrated that ferroptosis conditions affected mitochondrial structure and function, while the inhibitor Fer-1 restored mitochondrial structure and the inhibition of hypoxia-inducible factor 1α (HIF-1α) or the transferrin receptor 1 (TFRC) rescued IL-1β-induced loss of mitochondrial membrane potential. Inhibition of HIF-1α downregulated IL-1β-induced TFRC expression, while inhibition of TFRC did not downregulate IL-1β-induced HIF-1α expression in chondrocytes. Moreover, inhibition of HIF-1α or TFRC downregulated the IL-1β-induced MMP13 expression in chondrocytes, while inhibition of HIF-1α or TFRC rescued IL-1β-inhibited COL2A1 expression in chondrocytes. Furthermore, upregulation of TFRC promoted Fe2+ entry into chondrocytes, inducing the Fenton reaction and lipid peroxidation, which in turn caused ferroptosis, a disruption in chondrocyte functions, and an exacerbation of condylar cartilage degeneration. Together, these findings illustrate the far-reaching effects of chondrocyte ferroptosis in TMJOA as a mechanism causing chondrocyte death through iron overload, oxidative stress, and articular cartilage degeneration and a potential major cause of TMJOA.
Collapse
Affiliation(s)
- B Y Chen
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - J L Pathak
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - H Y Lin
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - W Q Guo
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - W J Chen
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - G Luo
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - L J Wang
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Vascular Biology Research Institute, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - X F Sun
- Department of Obstetrics and Gynecology; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Y Ding
- School of Medicine and Dentistry University of Rochester, Rochester, NY, USA
| | - J Li
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - T G H Diekwisch
- School of Medicine and Dentistry University of Rochester, Rochester, NY, USA
| | - C Liu
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Sun J, Song X, Wang C, Ruan Q. Geniposidic acid alleviates osteoarthritis progression through inhibiting inflammation and chondrocytes ferroptosis. J Cell Mol Med 2024; 28:e18228. [PMID: 38520209 PMCID: PMC10960175 DOI: 10.1111/jcmm.18228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/16/2024] [Accepted: 02/23/2024] [Indexed: 03/25/2024] Open
Abstract
Osteoarthritis is one of the common diseases that seriously affects the quality of life of middle-aged and elderly people worldwide. Geniposidic acid (GPA) is extracted from Eucommia ulmoides that exhibits various pharmacological effects. This study investigated the function of GPA on osteoarthritis (OA) in IL-1β-stimulated mouse chondrocytes and mouse OA model. Mouse OA model was established by destabilization of the medial meniscus (DMM) and GPA was given intraperitoneal injection. The results demonstrated that GPA could alleviate DMM-induced OA in mice. In vitro, IL-1β-induced PGE2, NO, MMP1 and MMP3 were suppressed by GPA. Furthermore, IL-1β-induced ferroptosis was inhibited by GPA, as confirmed by the inhibition of MDA, iron, and ROS, as well as the upregulation of GSH, GPX4, and Ferritin. In addition, GPA was found to increase the expression of Nrf2 and HO-1. And the inhibition of GPA on IL-1β-induced inflammation and ferroptosis were prevented by Nrf2 inhibitor. In conclusion, GPA alleviates OA progression through inhibiting inflammation and chondrocytes ferroptosis via Nrf2 signalling pathway.
Collapse
Affiliation(s)
- Jiayang Sun
- Department of OrthopedicsChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| | - Xianji Song
- Department of OrthopedicsChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| | - Cuijie Wang
- Department of AnesthesiologyChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| | - Qing Ruan
- Department of OrthopedicsChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
15
|
Lu Y, Lu D, Zhang H, Li H, Yu B, Zhang Y, Hu H, Sheng H. Causality between Ankylosing Spondylitis and osteoarthritis in European ancestry: a bidirectional Mendelian randomization study. Front Immunol 2024; 15:1297454. [PMID: 38380324 PMCID: PMC10876785 DOI: 10.3389/fimmu.2024.1297454] [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: 09/20/2023] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Objective To explore the bidirectional causal relationship between Ankylosing Spondylitis (AS) and Osteoarthritis (OA) at the genetic level within the European ancestry. Methods We implemented a series of quality control steps to select instrumental variables (IVs) related to the exposure. We conducted two-sample Mendelian randomization (MR) using the inverse-variance weighted method as the primary approach. We adjusted significance levels using Bonferroni correction, assessed heterogeneity using Cochrane's Q test. Sensitivity analysis was conducted through leave-one-out method. Additionally, external datasets and relaxed IV selection criteria were employed, and multivariate MR analyses were performed for validation purposes. Finally, Bayesian colocalization (COLOC) analysis identified common genes, validating the MR results. Results The investigation focused on the correlation between OA and AS in knee, hip, and hand joints. MR results revealed that individuals with AS exhibit a decreased risk of knee OA (OR = 0.9882, 95% CI: 0.9804-0.9962) but no significant increase in the risk of hip OA (OR = 0.9901, 95% CI: 0.9786-1.0018). Conversely, AS emerged as a risk factor for hand OA (OR = 1.0026, 95% CI: 1.0015-1.0036). In reverse-direction MR analysis, OA did not significantly influence the occurrence of AS. Importantly, minimal heterogeneity was observed in our MR analysis results (p > 0.05), and the robustness of these findings was confirmed through sensitivity analysis and multivariate MR analysis. COLOC analysis identified four colocalized variants for AS and hand OA (rs74707996, rs75240935, rs181468789, and rs748670681). Conclusion In European population, individuals with AS have a relatively lower risk of knee OA, whereas AS serves as a risk factor for hand OA. However, no significant causal relationship was found between AS and hip OA. Additionally, it offers novel insights into genetic research on AS and OA.
Collapse
Affiliation(s)
- Yangguang Lu
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Di Lu
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Hongzhi Zhang
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Haoyang Li
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Bohuai Yu
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Yige Zhang
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Hantao Hu
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Hongfeng Sheng
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
16
|
Pang N, Ding M, Yang H, Zhong Q, Zheng L, Luo D, Yao Y. Iron overload causes macrophages to produce a pro-inflammatory phenotype in the synovium of hemophiliac arthritis via the acetyl-p53 pathway. Haemophilia 2024; 30:195-203. [PMID: 38058260 DOI: 10.1111/hae.14905] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/09/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023]
Abstract
AIM Haemophiliac arthritis (HA) is caused by spontaneous intra-articular hemorrhage and repeated intra-articular hematomas, leading to iron overload, which, in turn, induces M1 macrophage polarisation and inflammatory cytokine secretion, resulting in synovitis. Here, we explored the mechanism by which iron overload in HA induces the polarisation of M1 macrophages, providing a new approach for the treatment of HA synovitis. METHODS The synovium from the knee joints of normal amputees and patients with HA was collected. Pathological changes in the synovial tissues were analysed using hematoxylin and eosin staining. Iron tissue deposition was evaluated using the iron assay kit and Prussia Blue staining, while macrophage phenotype was determined using immunofluorescence. The levels of pro-inflammatory cytokines and p53 acetylation were determine using western blotting. An in vitro iron overload model was established by inducing THP-1 macrophages with ferric ammonium citrate, and the involvement of acetylated p53 in M1 macrophage polarisation was investigated. RESULTS Compared to control samples, the iron content in the synovium of patients with HA was significantly increased. The protein levels of M1 macrophage markers, pro-inflammatory cytokines, and acetylated p53, were also significantly elevated in the synovial tissues of patients with HA. Similar results were observed in the in vitro iron overload model. Furthermore, the inhibition of p53 acetylation in vitro reversed these iron overload-induced effects. CONCLUSION In patients with HA, iron overload induced synovial p53 acetylation, leading to macrophage polarisation toward the M1 phenotype and increased inflammatory cytokine secretion, resulting in synovitis. HIGHLIGHTS Synovial iron overload is associated with changes in P53 acetylation in hemophiliac arthritis (HA). Acetylated p53, a known regulator of macrophage polarization, is highly expressed in HA synovium, suggesting a potential role in M1 polarization. HA synovial macrophages predominantly polarize into the pro-inflammatory M1 phenotype, secreting elevated levels of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Nanyu Pang
- Department of Orthopaedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingyang Ding
- Department of Orthopaedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hu Yang
- Department of Orthopaedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qigang Zhong
- Department of Orthopaedic Surgery, Ma Anshan People's Hospital, Ma Anshan, China
| | - Liujie Zheng
- Department of Orthopaedic Surgery, The Fourth Hospital of Wuhan, Wuhan, China
| | - Dasheng Luo
- Department of Orthopaedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yunfeng Yao
- Department of Orthopaedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
17
|
Huang Y, Chen L, Xiong B, Lu G, Chen C, Liu J. Integrating multiple microarray datasets to explore the significance of ferroptosis regulators in the diagnosis and subtype classification of osteoarthritis. Medicine (Baltimore) 2023; 102:e35917. [PMID: 37960823 PMCID: PMC10637513 DOI: 10.1097/md.0000000000035917] [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: 07/03/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease that reduces quality of life for patients. Ferroptosis plays a significant role in OA. However, its underlying mechanism remains unclear. In this study, we integrated 7 OA synovial datasets from the GEO database to screen for significant ferroptosis-related genes. The top 5 ferroptosis regulators were used to construct nomogram models to predict OA prevalence. Consensus clustering was applied to classify OA patients into different ferroptosis patterns based on significant ferroptosis-related genes. Subsequently, an immune cell infiltration study was performed to investigate the relationship between the significant ferroptosis regulators and immune cells. As a result, we screened 11 ferroptosis-related genes in OA patients. Five candidate ferroptosis regulators (SLC7A11, ALOX5, SLC1A5, GOT1, and GSS) were used to predict OA risk. The nomogram model based on these 5 genes is important for assessing the occurrence of OA. Consensus clustering analysis showed that OA patients could be classified into 2 ferroptosis patterns (Clusters A and B). Immune cell infiltration levels were higher in Cluster B than in Cluster A. Two subtypes, gene Clusters A and B, were classified according to the expression of ferroptosis-related DEGs among the ferroptosis patterns. Cluster A and gene Cluster A had higher ferroptosis scores than Cluster B or gene Cluster B, whereas the expression levels of the proinflammatory cytokines interleukin (IL)-1β, tumor necrosis factor, IL-6, IL-18, and IL-10 were higher in Cluster B or gene Cluster B than those in Cluster A or gene Cluster A. Different subtypes of ferroptosis play critical roles in OA. Furthermore, immunotherapy strategies for OA treatment may be guided by our study on ferroptosis patterns.
Collapse
Affiliation(s)
- Yue Huang
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - Lihua Chen
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - Bo Xiong
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - GuanYu Lu
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - Cai Chen
- First Clinical School of Medicine, Guangxi Traditional Chinese Medical University, Nanning, China
| | - JinFu Liu
- Department of Orthopedics and Traumatology, Xianhu District, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Guangxi, China
| |
Collapse
|
18
|
Sun J, Zhang Y, Wang C, Ruan Q. Kukoamine A protects mice against osteoarthritis by inhibiting chondrocyte inflammation and ferroptosis via SIRT1/GPX4 signaling pathway. Life Sci 2023; 332:122117. [PMID: 37741321 DOI: 10.1016/j.lfs.2023.122117] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
AIMS Osteoarthritis (OA) is one of the common chronic degenerative joint diseases, characterized by cartilage damage, subchondral bone changes, osteophyte formation, and synovitis. Kukoamine A (KuKA) is a bioactive compound isolated from Lycium chinense which is known as its anti-inflammatory activity. In this study, we detected the regulatory role of KuKA on OA both in vivo and in vitro. MATERIALS AND METHODS Mouse chondrocytes were cultured and mouse model of OA was established. Inflammatory mediator was measured by ELISA. The signaling pathway was tested by western blot analysis. KEY FINDINGS KuKA inhibited IL-1β-induced PGE2 and NO production and iNOS and COX-2 expression. IL-1β-induced MMP1 and MMP3 production was attenuated by KuKA. IL-1β-induced MDA, iron, and ROS were alleviated by KuKA. Meanwhile, GSH content, GPX4, Ferritin, SIRT1, Nrf2, and HO-1 expression were upregulated by KuKA. Furthermore, the inhibitory role of KuKA on IL-1β-induced inflammation, MMPs production, and ferroptosis were reversed by SIRT1 inhibitor. In vivo, KuKA could attenuate OA development in mouse model. KuKA markedly alleviated MMP1, MMP3, iNOS, and COX2 expression in OA mice. SIGNIFICANCE In conclusion, KuKA could inhibit OA development through suppressing chondrocyte inflammation and ferroptosis via SIRT1/GPX4 signaling pathway.
Collapse
Affiliation(s)
- Jiayang Sun
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Yunfeng Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Cuijie Wang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China
| | - Qing Ruan
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China.
| |
Collapse
|
19
|
Yang T, Yang G, Wang G, Jia D, Xiong B, Lu X, Li Y. Bioinformatics identification and integrative analysis of ferroptosis-related key lncRNAs in patients with osteoarthritis. Biosci Rep 2023; 43:BSR20230255. [PMID: 37702097 PMCID: PMC10500229 DOI: 10.1042/bsr20230255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/17/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Ferroptosis and dysregulation of long non-coding RNA (lncRNA) have been described to be strictly relevant to the pathogenesis of osteoarthritis (OA). However, the connection between ferroptosis and lncRNA in OA is poorly appreciated. Herein, we investigated the functional contribution of lncRNA markers correlated with the progression of human OA by comprehensive bioinformatics analysis of a panoramic network of competing endogenous RNA (ceRNA) based on ferroptosis-related genes (FRGs). METHODS FRGs-related competing endogenous RNA (ceRNA) networks were generated using differentially expressed genes based on OA-related whole transcriptome data from the Gene Expression Omnibus (GEO) database via starBase, miRTarBase, and miRWalk databases. The pivotal lncRNAs were ascertained by topological features (degree, betweenness, and closeness) and subceRNA networks were re-visualized. The expression difference of pivotal lncRNAs was verified by quantitative real-time polymerase chain reaction (qRT-PCR). The latent molecular mechanisms of the global ceRNA and subceRNA networks were uncovered by the R package clusterProfiler-based enrichment analysis. RESULTS A total of 98 dysregulated lncRNA-miRNA-mRNA regulatory relationships were attained in the FRGs-related panoramic ceRNA network of OA, covering 26 mRNAs, 20 miRNAs, and 20 lncRNAs. Three lncRNAs (AC011511.5, AL358072.1, and C9orf139) were ascertained as the central lncRNAs in the panoramic ceRNA network. Functional ensemble analysis illustrated that both the panoramic ceRNA network and the subceRNA network were integrally affiliated with the immune-inflammatory response, oxygen homeostasis, and cell death (apoptosis, autophagy, and ferroptosis). CONCLUSION Comprehensive bioinformatics analysis of the FRGs-related ceRNA network determined three molecular biomarkers of lncRNAs that might be affiliated with OA progression.
Collapse
Affiliation(s)
- Tengyun Yang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Guang Yang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Guoliang Wang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Di Jia
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Bohan Xiong
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Xiaojun Lu
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Yanlin Li
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| |
Collapse
|
20
|
Jin Z, Zhang H, Bai L, Yue L, Zhang W, Liang J, Chang B, Yang Y, Hu Z, Chen L, Guo C. Synovium is a sensitive tissue for mapping the negative effects of systemic iron overload in osteoarthritis: identification and validation of two potential targets. J Transl Med 2023; 21:661. [PMID: 37741987 PMCID: PMC10518090 DOI: 10.1186/s12967-023-04541-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/16/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND The prevention and treatment of osteoarthritis (OA) pose a major challenge in its research. The synovium is a critical tissue in the systematic treatment of OA. The present study aimed to investigate potential target genes and their correlation with iron overload in OA patients. METHODS The internal datasets for analysis included the microarray datasets GSE46750, GSE55457, and GSE56409, while the external datasets for validation included GSE12021 and GSE55235. The GSE176308 dataset was used to generate single-cell RNA sequencing profiles. To investigate the expression of the target genes in synovial samples, quantitative reverse transcription-PCR, western blotting, and immunohistochemical assay were conducted. ELISA was used to detect the levels of ferritin and Fe2+ in both serum and synovium. RESULTS JUN and ZFP36 were screened from the differentially expressed genes, and their mRNA were significantly reduced in the OA synovium compared to that in normal synovium. Subsequently, complex and dynamically evolving cellular components were observed in the OA synovium. The mRNA level of JUN and ZFP36 differed across various cell clusters of OA synovium and correlated with immune cell infiltration. Moreover, ferritin and Fe2+ were significantly increased in the serum and synovium of OA patients. Further, we found that JUN elevated and ZFP36 decreased at protein level. CONCLUSIONS The synovium is a sensitive tissue for mapping the adverse effects of systemic iron overload in OA. JUN and ZFP36 represent potential target genes for attenuating iron overload during OA treatment. Some discrepancies between the transcription and protein levels of JUN suggest that post-transcriptional modifications may be implicated. Future studies should also focus on the roles of JUN and ZFP36 in inducing changes in cellular components in the synovium during OA pathogenesis.
Collapse
Affiliation(s)
- Zhuangzhuang Jin
- Department of Emergence Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Zhang
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lingyu Yue
- Beijing AnDing Hospital of Capital Medical University, Beijing, China
| | - Weiming Zhang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiajian Liang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bohan Chang
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Yang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhehan Hu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liang Chen
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chuanji Guo
- Hospital Administration Office, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang City, Liaoning Province, China.
| |
Collapse
|
21
|
Guan Z, Jin X, Guan Z, Liu S, Tao K, Luo L. The gut microbiota metabolite capsiate regulate SLC2A1 expression by targeting HIF-1α to inhibit knee osteoarthritis-induced ferroptosis. Aging Cell 2023:e13807. [PMID: 36890785 DOI: 10.1111/acel.13807] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023] Open
Abstract
Ferroptosis is an iron-dependent cell death that has been found to aggravate the progression of osteoarthritis (OA) and gut microbiota- OA axis refers to the bidirectional information network between the gut microbiota and OA, which may provide a new way to protect the OA. However, the role of gut microbiota-derived metabolites in ferroptosis-relative osteoarthritis remains unclear. The objective of this study was to analyze the protective effect of gut microbiota and its metabolite capsiate (CAT) on ferroptosis-relative osteoarthritis in vivo and in vitro experiments. From June 2021 to February 2022, 78 patients were evaluated retrospectively and divided into two groups: The health group (n = 39) and the OA group (n = 40). Iron and oxidative stress indicators were determined in peripheral blood samples. And then in vivo and in vitro experiments, a surgically destabilized medial meniscus (DMM) mice model was established and treated with CAT or Ferric Inhibitor-1 (Fer-1). Solute Carrier Family 2 Member 1 (SLC2A1) short hairpin RNA (shRNA) was utilized to inhibit SLC2A1 expression. Serum iron was increased significantly but total iron binding capacity was decreased significantly in OA patients than healthy people (p < 0.0001). The least absolute shrinkage and selection operator clinical prediction model suggested that serum iron, total iron binding capacity, transferrin, and superoxide dismutase were all independent predictors of OA (p < 0.001). Bioinformatics results suggested that SLC2A1, Metastasis-Associated Lung Adenocarcinoma Transcript 1 (MALAT1), and HIF-1α (Hypoxia Inducible Factor 1 Alpha)-related oxidative stress signaling pathways play an important role in iron homeostasis and OA. In addition, gut microbiota 16s RNA sequencing and untargeted metabolomics were used to find that gut microbiota metabolites CAT in mice with osteoarthritis were negatively correlated with Osteoarthritis Research Society International (OARSI) scores for chondrogenic degeneration (p = 0.0017). Moreover, CAT reduced ferroptosis-dependent osteoarthritis in vivo and in vitro. However, the protective effect of CAT against ferroptosis-dependent osteoarthritis could be eliminated by silencing SLC2A1. SLC2A1 was upregulated but reduced the SLC2A1 and HIF-1α levels in the DMM group. HIF-1α, MALAT1, and apoptosis levels were increased after SLC2A1 knockout in chondrocyte cells (p = 0.0017). Finally, downregulation of SLC2A1 expression by Adeno-associated Virus (AAV) -SLC2A1 shRNA improves osteoarthritis in vivo. Our findings indicated that CAT inhibited HIF-1a expression and reduced ferroptosis-relative osteoarthritis progression by activating SLC2A1.
Collapse
Affiliation(s)
- Zhiyuan Guan
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China.,Department of Ophthalmology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Jin
- Department of Rheumatology and Immunology, The First People's Hospital of Xuzhou, Jiangsu, Xuzhou, China.,Department of Dermatology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Jiangsu, Xuzhou, China
| | - Zhiqiang Guan
- Department of Dermatology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Jiangsu, Xuzhou, China
| | - Shengfu Liu
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Kun Tao
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Liying Luo
- Department of Ophthalmology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Fighting age-related orthopedic diseases: focusing on ferroptosis. Bone Res 2023; 11:12. [PMID: 36854703 PMCID: PMC9975200 DOI: 10.1038/s41413-023-00247-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/03/2023] [Accepted: 01/11/2023] [Indexed: 03/02/2023] Open
Abstract
Ferroptosis, a unique type of cell death, is characterized by iron-dependent accumulation and lipid peroxidation. It is closely related to multiple biological processes, including iron metabolism, polyunsaturated fatty acid metabolism, and the biosynthesis of compounds with antioxidant activities, including glutathione. In the past 10 years, increasing evidence has indicated a potentially strong relationship between ferroptosis and the onset and progression of age-related orthopedic diseases, such as osteoporosis and osteoarthritis. Therefore, in-depth knowledge of the regulatory mechanisms of ferroptosis in age-related orthopedic diseases may help improve disease treatment and prevention. This review provides an overview of recent research on ferroptosis and its influences on bone and cartilage homeostasis. It begins with a brief overview of systemic iron metabolism and ferroptosis, particularly the potential mechanisms of ferroptosis. It presents a discussion on the role of ferroptosis in age-related orthopedic diseases, including promotion of bone loss and cartilage degradation and the inhibition of osteogenesis. Finally, it focuses on the future of targeting ferroptosis to treat age-related orthopedic diseases with the intention of inspiring further clinical research and the development of therapeutic strategies.
Collapse
|
23
|
Zhang F, Yan Y, Cai Y, Liang Q, Liu Y, Peng B, Xu Z, Liu W. Current insights into the functional roles of ferroptosis in musculoskeletal diseases and therapeutic implications. Front Cell Dev Biol 2023; 11:1112751. [PMID: 36819098 PMCID: PMC9936329 DOI: 10.3389/fcell.2023.1112751] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Ferroptosis is a novel type of cell death associated with iron accumulation and excessive lipid peroxidation. Elucidating the underlying molecular mechanisms of ferroptosis is intensively related to the development and treatment of multiple diseases, including musculoskeletal disorders. Moreover, in vitro and in vivo studies have shown the importance of oxidative stress in musculoskeletal conditions such as osteoporosis, osteoarthritis, rheumatoid arthritis, and osteosarcoma. Ferroptosis-derived clinical management of musculoskeletal diseases offers tremendous and attractive opportunities. Notably, ferroptosis agonists have been proven to enhance the sensitivity of osteosarcoma cells to conventional therapeutic strategies. In this review, we have mainly focused on the implications of ferroptosis regulation in the pathophysiology and therapeutic response of musculoskeletal disorders. Understanding roles of ferroptosis for controlling musculoskeletal diseases might provide directions for ferroptosis-driven therapies, which could be promising for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Cai
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Bi Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopedic Surgery, The Second Hospital University of South China, Hengyang, China
| |
Collapse
|
24
|
Theaflavin-3,3'-Digallate Inhibits Erastin-Induced Chondrocytes Ferroptosis via the Nrf2/GPX4 Signaling Pathway in Osteoarthritis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3531995. [PMID: 36439689 PMCID: PMC9691334 DOI: 10.1155/2022/3531995] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 11/19/2022]
Abstract
There is evidence that osteoarthritis (OA) is associated with ferroptosis which is a kind of lipid peroxidation-related cell death. Theaflavin-3,3'-digallate(TF3), a polyphenol compound extracted from black tea, possesses antioxidative and anti-inflammatory properties, but its effects on chondrocyte ferroptosis in osteoarthritis (OA) remain unclear. Our present study aims at exploring the protective role and underlying mechanisms of TF3 against erastin-induced chondrocyte ferroptosis in OA. In human primary chondrocytes treated with erastin alone or combined with different doses of TF3, cell viability was assessed by MTS. Ferroptosis-related proteins, including Gpx4, HO-1, and FTH1, were detected by western blot. The levels of lipid peroxidation and Fe2+ were determined by fluorescence staining. Meanwhile, the change of related proteins in the Nrf2/Gpx4 signaling pathway was determined by western blot. siRNA-mediated Nrf2 knockdown and the Gpx4 inhibitor RSL3 were used to explore molecular mechanisms for TF3-induced ferroptosis in OA chondrocyte. The magnetic resonance imaging (MRI), HE staining, Masson's staining, and immunohistochemistry were used to evaluate articular cartilage damages in the rat OA model. The results showed that Gpx4 expression was markedly downregulated in the chondrocytes of OA patients. TF3 reversed erastin-induced ferroptosis of human cultured chondrocytes, lipid ROS, and Fe2+ production in mitochondria. Moreover, the expression of Gpx4, HO-1, FTH1, and Nrf2 was markedly induced by TF3 in the erastin-treated chondrocytes. The antiferroptotic effect of TF3 was related to enhance Nrf2/Gpx4 signaling pathway. Finally, TF3 inhibited OA progression by alleviating in vivo cartilage damage related to chondrocyte ferroptosis. Thus, TF3 significantly inhibits chondrocyte ferroptosis by activating the Nrf2/Gpx4 signaling pathway, suggesting that TF3 serves as a potential therapeutic supplement for OA treatment.
Collapse
|
25
|
Genetic Causal Association between Iron Status and Osteoarthritis: A Two-Sample Mendelian Randomization. Nutrients 2022; 14:nu14183683. [PMID: 36145059 PMCID: PMC9501024 DOI: 10.3390/nu14183683] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Objective: Observational studies have shown the association between iron status and osteoarthritis (OA). However, due to difficulties of determining sequential temporality, their causal association is still elusive. Based on the summary data of genome-wide association studies (GWASs) of a large-scale population, this study explored the genetic causal association between iron status and OA. Methods: First, we took a series of quality control steps to select eligible instrumental SNPs which were strongly associated with exposure. The genetic causal association between iron status and OA was analyzed using the two-sample Mendelian randomization (MR). Inverse-variance weighted (IVW), MR-Egger, weighted median, simple mode, and weighted mode methods were used for analysis. The results were mainly based on IVW (random effects), followed by sensitivity analysis. IVW and MR-Egger were used for heterogeneity testing. MR-Egger was also used for pleiotropy testing. Leave-one-SNP-out analysis was used to identify single nucleotide polymorphisms (SNPs) with potential impact. Maximum likelihood, penalized weighted median, and IVW (fixed effects) were performed to further validate the reliability of results. Results: IVW results showed that transferrin saturation had a positive causal association with knee osteoarthritis (KOA), hip osteoarthritis (HOA) and KOA or HOA (p < 0.05, OR > 1), and there was a negative causal association between transferrin and HOA and KOA or HOA (p < 0.05, OR < 1). The results of heterogeneity test showed that our IVW analysis results were basically free of heterogeneity (p > 0.05). The results of the pleiotropy test showed that there was no pleiotropy in our IVW analysis (p > 0.05). The analysis results of maximum likelihood, penalized weighted median and IVW (fixed effects) were consistent with our IVW results. No genetic causal association was found between serum iron and ferritin and OA. Conclusions: This study provides evidence of the causal association between iron status and OA, which provides novel insights to the genetic research of OA.
Collapse
|
26
|
Pain in Hemophilia: Unexplored Role of Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11061113. [PMID: 35740010 PMCID: PMC9220316 DOI: 10.3390/antiox11061113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 02/04/2023] Open
Abstract
Hemophilia is the most common X-linked bleeding diathesis caused by the genetic deficiency of coagulation factors VIII or IX. Despite treatment advances and improvements in clinical management to prevent bleeding, management of acute and chronic pain remains to be established. Repeated bleeding of the joints leads to arthropathy, causing pain in hemophilia. However, mechanisms underlying the pathogenesis of pain in hemophilia remain underexamined. Herein, we describe the novel perspectives on the role for oxidative stress in the periphery and the central nervous system that may contribute to pain in hemophilia. Specifically, we cross examine preclinical and clinical studies that address the contribution of oxidative stress in hemophilia and related diseases that affect synovial tissue to induce acute and potentially chronic pain. This understanding would help provide potential treatable targets using antioxidants to ameliorate pain in hemophilia.
Collapse
|
27
|
Wu L, Si H, Zeng Y, Wu Y, Li M, Liu Y, Shen B. Association between Iron Intake and Progression of Knee Osteoarthritis. Nutrients 2022; 14:nu14081674. [PMID: 35458236 PMCID: PMC9033045 DOI: 10.3390/nu14081674] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Iron overload is drawing attention in the development of knee osteoarthritis (OA). To identify the modifiable risk factors for iron-related pathological conditions, we examined the association between iron intake and the risk of knee OA progression. Methods: A total of 1912 participants in the Osteoarthritis Initiative (OAI), aged 45−79 years and with at least one knee radiographic OA at baseline, were identified and were followed up to 6 years. The iron and other nutrient intake was measured by the validated Block Brief 2000 Food Frequency Questionnaire. The outcome measures were by radiographic progression on the basis of the Kellgren−Lawrence (KL) grade and the joint-space-narrowing (JSN) score. The association between the iron intake and the knee OA progression was examined by Cox proportional hazards models and restricted cubic spline (RCS) regression. Results: Among the study participants, 409 participants experienced KL-grade progression, and 684 participants experienced JSN-score progression within 6 years. Overall, the association between iron intake and the risk of KL-grade progression followed a U shape (p for nonlinearity < 0.001). The risk of KL-grade progression was significantly lower in participants with iron intakes of <16.5 mg/day (per mg/day: adjusted hazard ratio (HR), 0.75; 95% CI (confidence interval), 0.64−0.89), and it was higher in those with iron intakes ≥16.5 mg/day (per mg/day: HR, 1.20; 95% CI, 1.04−1.38). Consistently, when the iron intake was assessed as deciles, compared to those in Deciles 3−5 (10.9−23.3 mg/day), the risk of KL-grade progression was higher for Deciles 1−2 (≤10.9 mg/day: HR, 1.57; 95% CI, 1.17−2.10) and for Deciles 6−10 (>23.3 mg/day: adjusted HR, 1.60; 95% CI, 1.19−2.16). Similar U-shaped relations were found for iron intake with the risk of JSN-score progression (p for nonlinearity = 0.035). Conclusions: There was a U-shaped association between the iron intake and the progression of knee OA, with an inflection point at about 16.5 mg/day, and minimal risk from 10.9 to 23.3 mg/day of iron intake. An appropriate iron intake was advisable for knee OA, whereas excessive or deficient iron intake increased the risk of knee OA progression.
Collapse
|