1
|
Zheng G, Lu M, Ouyang Y, Sun G. RNA methylation: A new perspective in osteoarthritis research. Gene 2025; 959:149518. [PMID: 40254081 DOI: 10.1016/j.gene.2025.149518] [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: 02/04/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by cartilage degradation, osteophyte formation, and joint dysfunction, significantly impairing the quality of life in the elderly population. Recently, RNA modifications, as a dynamic and reversible epigenetic modification, have emerged as critical players in the onset and progression of OA. This review systematically summarizes the major types of RNA modifications involved in OA, including N6-methyladenosine (m6A), 5-methylcytosine (m5C), and 7-methylguanosine (m7G), and explores their roles in regulating chondrocyte autophagy, inflammatory responses, and key signaling pathways. with a primary focus on RNA methylation. Special emphasis is placed on the dynamic regulatory functions of key methyltransferases (e.g., METTL3, FTO, WTAP) and their potential contributions to OA pathogenesis. Furthermore, we address current research hotspots and controversies in the field, proposing future research directions, such as leveraging single-cell sequencing to decipher dynamic RNA modification changes during OA progression and uncovering the cooperative networks among various RNA modifications. Advancing our understanding of the biological roles and mechanisms of RNA modifications holds promise for innovative strategies in the early diagnosis, disease stratification, and targeted therapy of OA.
Collapse
Affiliation(s)
- Guihao Zheng
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China; Graduate School of Jiangxi Medical College, Nanchang University, China.
| | - Meifeng Lu
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China; Graduate School of Jiangxi Medical College, Nanchang University, China.
| | - Yulong Ouyang
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China.
| | - Guicai Sun
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China.
| |
Collapse
|
2
|
Li Q, Li C, Liu X, Guo Z, Li X, Zhang X. The key role of Piezo1 channels in ferroptosis after spinal cord injury and the therapeutic potential of Piezo1 inhibitors. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:132-140. [PMID: 40339662 DOI: 10.1016/j.pbiomolbio.2025.05.001] [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: 02/03/2025] [Revised: 04/07/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Ferroptosis has been confirmed to be one of the key mechanisms of neuronal injury and dysfunction after spinal cord injury (SCI). Mechanical stresses such as deformation, compression, and stretching not only directly cause physical damage to spinal cord tissue at the moment of SCI, but also promote the development of ferroptosis through various pathways. However, the mechanism of ferroptosis after SCI remains unclear, which hinders the development of therapeutic methods. OBJECTIVE This article aims to review the key mechanisms by which mechanical stress affects ferroptosis after SCI, including its impact on the structure and function of the endoplasmic reticulum (ER) and mitochondria, its role in triggering inflammatory responses, and its activation of mechanosensitive channels. Special emphasis is placed on the role of Piezo1 channels, which are key factors in cell mechanosensation and ion homeostasis regulation. The review explores how Piezo1 channels are upregulated by mechanical stress after SCI and participate in the ferroptosis process by mediating ion flow and other mechanisms. CONCLUSIONS Inhibiting Piezo1 channels may be a potential therapeutic strategy for SCI. This review summarizes the therapeutic potential of Piezo1 inhibitors by sorting out existing studies, hoping to provide a theoretical basis for effective therapeutic strategies targeting ferroptosis after SCI.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinyu Liu
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Zixuan Guo
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinxin Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
3
|
Shinohara I, Morita M, Chow SKH, Murayama M, Sususki Y, Gao Q, Goodman SB. Pathophysiology of the Effects of Oxidative Stress on the Skeletal System. J Orthop Res 2025; 43:1059-1072. [PMID: 40143581 DOI: 10.1002/jor.26075] [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: 01/29/2025] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025]
Abstract
Reactive oxygen species (ROS) are molecules that are generated primarily during energy production in cells. ROS are involved in critical biological functions such as signal transduction; when the production of ROS is imbalanced, excessive ROS causes oxidative stress, and subsequent cellular damage. Oxidative stress is linked to numerous pathological disorders in major organs including the skeletal system. In an aging society, understanding the role of ROS in skeletal health is critical to developing preventative and therapeutic interventions. Oxidative stress causes defects in cellular differentiation, apoptosis, mitochondrial dysfunction, and inflammation. The effects of oxidative stress on the skeletal system have been implicated in the development of osteoporosis, knee osteoarthritis, and osteonecrosis by inhibiting bone remodeling, increasing osteoclast activity, and decreasing osteoblast function. ROS are also involved in many signaling pathways that regulate immune defense, cell proliferation, and inflammation. This underscores the importance of maintaining a balance between ROS and antioxidants to prevent oxidative stress and related diseases. Targeting ROS and oxidative stress mechanisms may offer new treatments for diseases affecting the skeletal system and other organs, potentially improving health outcomes, and extending healthy lifespans. This review highlights the significant impact of oxidative stress on skeletal health and explores potential preventative and therapeutic strategies to mitigate the adverse effects of ROS.
Collapse
Affiliation(s)
- Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Mayu Morita
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yosuke Sususki
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
4
|
Yi G, Li M, Zhou J, Li J, Song X, Li S, Liu J, Zhang H, Chen Z. Novel pH-responsive lipid nanoparticles deliver UA-mediated mitophagy and ferroptosis for osteoarthritis treatment. Mater Today Bio 2025; 32:101697. [PMID: 40225130 PMCID: PMC11986606 DOI: 10.1016/j.mtbio.2025.101697] [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: 10/14/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Synovial inflammation plays a crucial role in osteoarthritis (OA) development, leading to chronic inflammation and cartilage destruction. Although targeting synovitis can alleviate OA, clinical outcomes have been disappointing due to poor drug targeting and joint cavity heterogeneity. This study presents pH-responsive lipid nanoparticles (LNPs@UA), loaded with Urolithin A (UA), as a potential OA treatment. LNPs@UA showed uniform particle size, low zeta potential, and effective mitochondria-targeting and pH-responsive capabilities. In vitro, LNPs@UA reduced reactive oxygen species (ROS), pro-inflammatory factors (IL-1β, IL-6, TNF-α), and promoted M2 macrophage polarization. It improved mitochondrial structure, enhanced autophagy, and inhibited ferroptosis. In vivo, LNPs@UA alleviated OA progression in an ACLT-induced OA mouse model. Transcriptomic analysis revealed inhibition of NF-κB signaling and activation of repair pathways. These results suggest LNPs@UA could offer a promising therapeutic approach for OA.
Collapse
Affiliation(s)
- Guoliang Yi
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Guizhou Medical University, Guiyang, 550004, China
| | - Min Li
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiayi Zhou
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jinxin Li
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xizheng Song
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Siming Li
- Guizhou Medical University, Guiyang, 550004, China
- Department of Orthopedics, Guangzhou Red Cross Hospital, Guangzhou, 510220, China
| | - Jianghua Liu
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haowei Zhang
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zhiwei Chen
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
5
|
Liu Y, Wang Y, Cheng S, Mu J, Yin G, Gao H. Pantothenic acid alleviates osteoarthritis progression by inhibiting inflammatory response and ferroptosis through the SIRT1/Nrf2 signaling pathway. Chem Biol Interact 2025; 413:111494. [PMID: 40157627 DOI: 10.1016/j.cbi.2025.111494] [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: 01/04/2025] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Osteoarthritis (OA) is a degenerative joint disease that is a major cause of deformity, swelling, pain and even loss of function in the knee joints of the elderly. Pantothenic acid (PA) plays a protective role in many organs due to its antioxidant and anti-inflammatory properties. Herein, we aimed to assess the protective role of PA on osteoarthritis and investigate the underlying molecular mechanism. The levels of inflammatory factors (IL-1β and TNF-α) in knee tissues were measured by ELISA. The Safranin O-Fast Green staining was used to assess the severity of OA and the H&E staining was used to assess the degree of synovitis. In vitro, the levels of iron, MDA, GSH were measured by the detection kits. Western blotting was used to assess the levels of signaling-related proteins. Our results showed that PA significantly attenuated the degree of cartilage degeneration in the MIA-induced osteoarthritis model. PA also reduced the expression of IL-1β, TNF-α, MMP1 and MMP3. In vitro, PA effectively reduced the concentrations of MMP1 and MMP3 in IL-1β-stimulated chondrocytes. PA decreased the levels of Fe2+ and MDA, while increasing GSH production and GPX4 and SLC7A11 expression in IL-1β-induced chondrocytes. Meanwhile, we found that PA was able to inhibit the phosphorylation level of p65, IκB protein in chondrocytes, which effectively blocked the NF-κB signaling pathway. Furthermore, PA also increased the level of SIRT1, Nrf2, and HO-1 protein expression. In addition, the inhibition of PA on IL-1β-induced MMPs production and ferroptosis were inhibited by the SIRT1 inhibitor EX-527. In conclusion, PA inhibited chondrocyte ferroptosis and cartilage destruction in osteoarthritis. The mechanism was through activating SIRT1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yi Liu
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yang Wang
- The First Operation Room, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shengqi Cheng
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jie Mu
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Guanchen Yin
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Hang Gao
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
6
|
Zhang Y, Zhao CY, Zhou Z, Li CC, Wang Q. The effect of lactate dehydrogenase B and its mediated histone lactylation on chondrocyte ferroptosis during osteoarthritis. J Orthop Surg Res 2025; 20:493. [PMID: 40394653 PMCID: PMC12093889 DOI: 10.1186/s13018-025-05894-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 05/06/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Histone lactylation is a novel epigenetic regulator that is reported to participate in gene expression. Ferroptosis is an oxidative form of cell death and chondrocyte ferroptosis crucially impacts the development of osteoarthritis (OA). The study aimed at investigating the effect of lactate dehydrogenase B (LDHB) and its mediated histone lactylation on chondrocyte ferroptosis during OA. METHODS Our study focused on the establishment of in vivo mouse model and in vitro interleukin-1β (IL-1β)-induced chondrocytes model and administrated LDHB knockdown (siLDHB). Histopathological assessment of cartilage was conducted via HE staining, while serum levels of cartilage oligomeric matrix protein (COMP) and crosslinked C-telopeptides of type II collagen (CTX-II) were quantified using ELISA to evaluate OA severity. The matrix degradation was further examined by expression of Collagen II and Aggrecan. Levels of total iron, ferrous iron (Fe2+), and lipid reactive oxygen species (ROS) were considered measurements of ferroptosis. Assessment of cell viability and proliferation relied on cell counting kit 8 (CCK-8) together with colony formation assay. Western blotting assay served for detecting the relative expression of proteins and protein lactylation. The epigenetic regulation of ACSL4 by LDHB was determined by chromatin immunoprecipitation (ChIP) and luciferase reporter gene assay. RESULTS OA mice presented remarkably elevated protein level of LDHB and H3K18 lactylation in the cartilage versus the sham group. Knockdown of LDHB downregulated the levels of COMP and CTX-II, as well as alleviated chondrocyte ferroptosis in vitro and in vivo. Results from ChIP and luciferase reporter gene assay demonstrated direct histone lactylation of ACSL promoter, and knockdown of LDHB and treatment with LDH inhibitor reduced histone lactylation and expression of ACSL4. ACSL4 overexpression could reverse the impact of LDHB depletion on chondrocyte proliferation and ferroptosis. CONCLUSION LDHB promotes ACSL4 by histone lactylation to induce chondrocyte ferroptosis, which further contributes to OA development. The findings in the study assist in understanding the modulating mechanism of LDHB-mediated lactylation against chondrocyte ferroptosis in OA progression.
Collapse
Affiliation(s)
- Yang Zhang
- Dalian Medical University, No.9, West Section of Lushun South Road, Dalian, 116044, Liaoning Province, China
| | - Chen-Yu Zhao
- Department of Orthopedics, The Third People's Hospital of Dalian, Non-Directly Affiiated Hospital of Dalian Medical University, No.40, Qianshan Road, Dalian, 116091, Liaoning, China
| | - Zheng Zhou
- Department of Orthopedics, Yangzhou Hongquan Hospital, Yangzhou University Medical College, Longchuan North Road, Jiangdu District, Yangzhou City, 225200, Jiangsu Province, China
| | - Cheng-Cun Li
- Department of Orthopedics, Yangzhou Hongquan Hospital, Yangzhou University Medical College, Longchuan North Road, Jiangdu District, Yangzhou City, 225200, Jiangsu Province, China
| | - Qiang Wang
- Dalian Medical University, No.9, West Section of Lushun South Road, Dalian, 116044, Liaoning Province, China.
- Yangzhou Clinical College of Medicine, Dalian Medical University, No. 98, Nantong West Road, Wenhe Street, Yangzhou City, 225009, Jiangsu Province, China.
| |
Collapse
|
7
|
Li W, Liu Y, Wei M, Yang Z, Tang H, Huang W. Chondrocyte-targeted α-Solanine through HIF-1α regulating glycolysis to reduce the ferroptosis of chondrocyte in osteoarthritis. Int Immunopharmacol 2025; 159:114841. [PMID: 40394792 DOI: 10.1016/j.intimp.2025.114841] [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: 02/23/2025] [Revised: 05/03/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
α-Solanine, a glycoalkaloid (GA) extracted from the stems of the potato plant, exhibits bioactivity and medicinal potential that necessitate further investigation. The impact and underlying mechanisms of α-Solanine on osteoarthritis (OA) remain to be elucidated. To achieve enhanced therapeutic outcomes, we have designed and synthesized a UIO-66-NH2@α-Solanine@PEI charged particle (USP) that amplifies the therapeutic effects of α-Solanine, demonstrating superior efficacy. Our approach involved the synthesis of a novel drug delivery system, the USP, to augment the therapeutic potential of α-Solanine in the treatment of OA. An OA rat model was established, and USP treatment was administered. The therapeutic effects were verified through histochemical staining and micro-CT. In vitro, α-Solanine significantly suppressed the expression of proteins related to glycolysis and notably inhibited ferroptosis. RNA sequencing revealed hypoxia-inducible factor-1α (HIF-1α) as a potential pathway mediating the effects of α-Solanine, and it was found that the co-addition of cycloheximide (CHX) led to a shortened decay time of HIF-1α. In vivo, rats with OA demonstrated significant inhibition of glycolysis and ferroptosis following treatment with USP, along with improvements in OA characteristics. These findings suggest that α-Solanine can inhibit the intense glycolysis associated with OA via the HIF-1α pathway and alleviate ferroptosis in chondrocytes. Treatment with USP demonstrated superior efficacy in the management of OA, providing a new therapeutic strategy for the disease.
Collapse
Affiliation(s)
- Wenwei Li
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232000, China; Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yang Liu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Ming Wei
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Zhichao Yang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Hao Tang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Wei Huang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
8
|
Ye QH, Zhang P, Zhao YH, Zhu WX, Zhu HX, Wei BF. Decreased serum and local GPX4 and SLC7A11 expression correlates with disease severity in non-traumatic osteonecrosis of the femoral head. J Orthop Surg Res 2025; 20:477. [PMID: 40380264 PMCID: PMC12084951 DOI: 10.1186/s13018-025-05912-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 05/11/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Ferroptosis is implicated in various musculoskeletal conditions, including non-traumatic osteonecrosis of the femoral head (NT-ONFH). OBJECTIVE The objective of this study was to explore the levels of two crucial proteins associated with ferroptosis, namely Glutathione peroxidase 4 (GPX4) and Solute Carrier Family 7 Member 11 (SLC7A11), in both serum and femoral head samples, and to correlate their expression levels with the clinical severity of NT-ONFH. METHODS The study included 136 NT-ONFH patients and an equal number of healthy controls. In addition, 68 subjects suffering from femoral neck fractures (FNF) were included in the study. The serum concentrations of GPX4 and SLC7A11 were quantified using the enzyme-linked immunosorbent assay. The GPX4 and SLC7A11 levels among tissue samples were identified through immunohistochemical staining, western blot analysis, and quantitative real-time polymerase chain reaction (qRT-PCR). The radiographic severity of the condition was evaluated utilizing the Association Research Circulation Osseous (ARCO) classification system, while the symptomatic severity was assessed utilizing the Visual Analogue Scale (VAS) alongside the Harris Hip Score (HHS). RESULTS Patients diagnosed with NT-ONFH had considerably reduced serum concentrations of GPX4 and SLC7A11 in comparison to individuals in the healthy control group. Negative correlations of serum GPX4 and SLC7A11 levels with the ARCO stages were observed. A total of 73 ONFH and 68 FNF patients underwent total hip replacement. The mRNA and protein levels of GPX4 and SLC7A11 were lower in the necrotic areas compared to the non-necrotic areas and FNF femoral head tissues. Subsequent Receiver operating characteristic (ROC) curve analysis suggested that the decreased levels of both serum and local GPX4 and SLC7A11 could serve as potential biomarkers for the progression of ONFH. Furthermore, serum and local GPX4 and SLC7A11 levels were found to be negatively linked to the VAS score but positively related to the HHS score. CONCLUSION The levels of GPX4 and SLC7A11, both in serum and at the local site, were inversely correlated with the progression of NT-ONFH. Targeting ferroptosis and its associated proteins through potential therapeutic interventions could be a viable strategy to mitigate the severity of NT-ONFH.
Collapse
Affiliation(s)
- Qing-He Ye
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong Province, China
- Linyi People's Hospital postgraduate training base of Guangzhou University of Traditional Chinese Medicine, Linyi, Shandong Province, China
| | - Peng Zhang
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong Province, China
- Linyi People's Hospital postgraduate training base of Guangzhou University of Traditional Chinese Medicine, Linyi, Shandong Province, China
| | - Yong-Heng Zhao
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong Province, China
- Linyi People's Hospital postgraduate training base of Guangzhou University of Traditional Chinese Medicine, Linyi, Shandong Province, China
| | - Wen-Xiu Zhu
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong Province, China
- Linyi People's Hospital postgraduate training base of Guangzhou University of Traditional Chinese Medicine, Linyi, Shandong Province, China
| | - Hong-Xun Zhu
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong Province, China
- Linyi People's Hospital postgraduate training base of Guangzhou University of Traditional Chinese Medicine, Linyi, Shandong Province, China
| | - Biao-Fang Wei
- Linyi People's Hospital postgraduate training base of Guangzhou University of Traditional Chinese Medicine, Linyi, Shandong Province, China.
| |
Collapse
|
9
|
Wang T, Liu X, Feng X, Zhang Z, Lv R, Feng W, Zhou Y, Liao X, Tang H, Xu M. GPX4 degradation contributes to heat stress-induced liver injury via chaperone-mediated autophagy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119988. [PMID: 40368268 DOI: 10.1016/j.bbamcr.2025.119988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/19/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Heat stress (HS) is a significant health concern that adversely affects both human and animal health, particularly impacting liver function due to its central metabolic role. This study investigated the mechanisms underlying HS-induced liver injury, focusing on the role of ferroptosis, an iron-dependent form of cell death characterized by lipid peroxidation and cellular iron accumulation. Using mouse and cellular HS models, the results demonstrated that HS induced liver injury through ferroptosis, as evidenced by increased levels of malondialdehyde (MDA), oxidized glutathione (GSSG), and iron, alongside decreased glutathione (GSH) and glutathione peroxidase 4 (GPX4) expression. The ferroptosis inhibitor Ferrostatin-1 (Fer-1) effectively mitigated HS-induced liver damage, reducing oxidative stress and restoring GPX4 levels. Furthermore, HS promoted the lysosomal degradation of GPX4 via the chaperone-mediated autophagy (CMA) pathway, which was regulated by heat shock cognate protein 70 (HSC70) and lysosome-associated membrane protein 2A (LAMP2A). Knockdown of LAMP2A in hepatocytes significantly suppressed HS-induced GPX4 degradation, confirming the critical role of CMA in this process. Inhibition of CMA using Apoptozole, an HSC70 inhibitor, or Bafilomycin A1 (Baf-A1), a lysosomal inhibitor, further attenuated HS-induced ferroptosis and liver injury. These findings highlight the critical role of CMA-mediated GPX4 degradation in HS-induced ferroptosis and liver injury, providing potential therapeutic targets for mitigating HS-related liver damage.
Collapse
Affiliation(s)
- Ting Wang
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xiao Liu
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xinyu Feng
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Zhenyu Zhang
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ruiyi Lv
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Wenhong Feng
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yukun Zhou
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xueyu Liao
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Haoming Tang
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ming Xu
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.
| |
Collapse
|
10
|
Zhang Y, Wu Y, Ding B, Li Q, Chen X, Liu H, Xu M, Lan Y, Li Y. TNF-α inhibits Epstein Barr virus reactivation through the GPX4 mediated glutathione pathway. Sci Rep 2025; 15:16448. [PMID: 40355596 PMCID: PMC12069636 DOI: 10.1038/s41598-025-98679-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 04/14/2025] [Indexed: 05/14/2025] Open
Abstract
Epstein-Barr virus (EBV) is a carcinogenic γ-herpesvirus that remains latent in more than 95% of adults. The virus can undergo lytic activation when immune function is suppressed or when stimulated by drugs or pathogens. EBV reactivation poses a significant threat to human health and is closely associated with various cancers, such as Burkitt's lymphoma and nasopharyngeal carcinoma. Inhibiting EBV reactivation is a current clinical challenge. Tumour necrosis factor-α (TNF-α), an important cytokine, has different effects on various viruses. It also exerts varying effects on the same virus depending on the type of infected cell. This study aimed to investigate the impact of TNF-α on EBV reactivation and its underlying mechanisms. Our experimental research revealed that TNF-α significantly inhibits EBV reactivation and that this inhibitory effect is mediated primarily through its receptor TNFR1. Furthermore, TNF-α affects the expression of the GPX4 protein and regulates the potential ferroptosis state of cells. Using transmission electron microscopy and other methods, we observed typical characteristics of ferroptosis, such as changes in mitochondrial morphology and Fe2 + accumulation. Additionally, we established stable GPX4-knockdown cell lines, which demonstrated the crucial role of GPX4 in the process of TNF-α-mediated inhibition of EBV reactivation. Overall, TNF-α acts on the TNFR1 receptor, thereby affecting the GPX4 protein and the ferroptosis pathway to achieve its inhibitory effect on EBV reactivation. These findings provide new insights into the mechanisms of EBV reactivation and may offer new perspectives for the early treatment of EBV-related diseases.
Collapse
Affiliation(s)
- Youyu Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yilin Wu
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Beining Ding
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qian Li
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xuenuo Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Huiling Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Mingyan Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yinghua Lan
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Yongguo Li
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
11
|
Yang J, Jiang H, Wu C, Lin Y, Tan G, Zhan J, Han L, Zhu Y, Shang P, Liu L, Liu H. Copper silicate nanoparticle-mediated delivery of astragaloside-IV for osteoarthritis treatment by remodeling the articular cartilage microenvironment. J Control Release 2025; 381:113583. [PMID: 40032006 DOI: 10.1016/j.jconrel.2025.113583] [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: 10/28/2024] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
With the increasing global aging population, osteoarthritis (OA) has emerged as a major public health concern. OA pathogenesis is characterized by a complex interplay among inflammatory cytokines, reactive oxygen species, and extracellular matrix components, leading to cartilage degradation. Astragaloside-IV (AS-IV), a natural antioxidant, has shown promise in alleviating OA symptoms but is limited by poor bioavailability and ineffective cartilage drug delivery. To address these challenges, we aimed to develop a drug delivery system using copper silicate nanoparticles modified with polyethylene glycol and loaded with AS-IV (referred to as CSP@AS-IV). This system uses mesoporous silica nanoparticles with a hybrid metal framework to enhance drug release and efficacy. CSP@AS-IV degrades in the acidic OA microenvironment, releasing copper ions (Cu2+) and AS-IV, which synergistically exert antioxidant, antibacterial, anti-inflammatory, and chondroprotective effects. Both in vitro and in vivo rat model experiments demonstrated that CSP@AS-IV significantly alleviated joint inflammation, downregulated inflammatory marker expression, and promoted cartilage repair. These findings underscore that CSP@AS-IV offers considerable clinical potential for enhancing OA treatment outcomes.
Collapse
Affiliation(s)
- Jianfeng Yang
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hongyi Jiang
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Congcong Wu
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuzhe Lin
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Guancan Tan
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Juannan Zhan
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Lijiang Han
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yiting Zhu
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ping Shang
- Department of Rehabilitation, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Haixiao Liu
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
12
|
Li Y, Liao X, Yu X, Xiao Y, Tao X, Zhong T. Mediating Role of the ANGPTL3/TFPI Protein Ratio in Regulating T-Cell Surface Glycoprotein CD5 Levels on Knee Osteoarthritis (KOA): A Mendelian Randomization Study. Int J Mol Sci 2025; 26:4471. [PMID: 40429617 DOI: 10.3390/ijms26104471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
This study utilized Mendelian randomization (MR) to investigate the impact of inflammatory proteins on knee osteoarthritis (KOA), measured using the ratio of protein levels (rQTLs). The primary objective was to identify potential intervention targets to mitigate KOA progression. Data from 2821 rQTLs, 91 inflammatory proteins, and KOA-related genetic variations were obtained through genome-wide association studies (GWAS). Bidirectional MR identified rQTLs with unidirectional causal relationships with KOA. Further analyses included false discovery rate (FDR) correction, colocalization, and mediation analysis. Two inflammatory proteins were found to be associated with KOA: T-cell surface glycoprotein CD5 [OR (95% CI) = 0.867 (0.760-0.990), PIVW = 0.035] and C-X-C motif chemokine 9 [OR (95% CI) = 1.150 (1.001-1.320), PIVW = 0.048]. Variations in their levels influenced rQTLs, producing differential effects on KOA. Specifically, rQTL-ANGPTL3/TFPI (human recombinant angiopoietin-like protein 3/Tissue factor pathway inhibitor) was identified as a mediator in the effect of T-cell surface glycoprotein CD5 levels on KOA. T-cell surface glycoprotein CD5 levels were negatively correlated with rQTL-ANGPTL3/TFPI (β1 = -0.084), while rQTL-ANGPTL3/TFPI was positively correlated with KOA (β2 = 0.159). These findings align with the total effect, where T-cell surface glycoprotein CD5 levels were negatively associated with KOA (β = -0.143). Thus, rQTL-ANGPTL3/TFPI may serve as a reliable mediator in the pathway through which T-cell surface glycoprotein CD5 levels affect KOA. This mediator may not only represent a potential therapeutic target but also serve as a biomarker for assessing KOA treatment efficacy, offering a novel direction for KOA diagnosis and management.
Collapse
Affiliation(s)
- Yongwei Li
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Xi Liao
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Xi Yu
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Ying Xiao
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Xiaoyu Tao
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Tian Zhong
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| |
Collapse
|
13
|
Liu Y, Wang Q, Hou Z, Gao Y, Li P. Electroacupuncture Inhibits Ferroptosis by Modulating Iron Metabolism and Oxidative Stress to Alleviate Cerebral Ischemia-Reperfusion Injury. J Mol Neurosci 2025; 75:63. [PMID: 40317390 PMCID: PMC12049298 DOI: 10.1007/s12031-025-02355-2] [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: 02/19/2025] [Accepted: 04/19/2025] [Indexed: 05/07/2025]
Abstract
Ischemic stroke (IS) is one of the leading causes of mortality and long-term disability worldwide. Electroacupuncture (EA) is commonly used in the treatment of IS, meaning that may reduce cerebral ischemia-reperfusion injury (CIRI). The middle cerebral artery occlusion/reperfusion (MCAO/R) rat models were created by the modified Zea Longa suture method. EA treatment was performed for 7 consecutive days at the acupoints Neiguan (PC6), Shuigou (GV26), and Sanyinjiao (SP6). The neurological function was assessed using the Zausinger six-point neurological deficiency score. The cerebral infarct volume was detected by 2,3,5-triphenyl tetrazolium chloride (TTC) staining. Hematoxylin and eosin (HE) staining was employed to observe the pathological changes in brain tissues. Prussian blue staining was employed to investigate iron deposition within the brain tissues. Transmission electron microscopy (TEM) was utilized to examine the morphological characteristics of mitochondria. Simultaneously, flow cytometry was utilized to detect the fluorescence intensity of reactive oxygen species (ROS). Assay kits were employed to measure the levels of Fe2+ and glutathione (GSH). Additionally, western blot (WB) and real-time quantitative polymerase chain reaction (RT-qPCR) assays were performed to evaluate the expression levels of proteins associated with ferroptosis. Compared with the MCAO/R group, both the MCAO/R + EA and MCAO/R + DFO groups exhibited significant improvements in neurological function following cerebral ischemia-reperfusion (CIR), attenuated the pathological brain tissue injury, and reduced the cerebral infarct volume and iron deposition in brain tissue. Furthermore, both the MCAO/R + EA and MCAO/R + DFO groups displayed a marked reduction in mitochondrial injury. There was a substantial decrease in Fe2+ and ROS levels, accompanied by a notable increase in GSH level and glutathione peroxidase 4 (GPX4) activity. Compared with the MCAO/R group, the levels of ferroportin1 (FPN1) protein and mRNA expression were significantly increased in the MCAO/R + EA and MCAO/R + DFO groups, and the expression levels of transferrin (TF), transferrin receptor 1 (TFR1), divalent metal transporter 1 (DMT1) protein and mRNA, as well as ferritin (FER) protein, were significantly decreased. EA inhibits ferroptosis by modulating iron metabolism and oxidative stress to alleviate CIRI, exerting neuroprotective effects.
Collapse
Affiliation(s)
- Yaoyao Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qi Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Ziwen Hou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Ying Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Peng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
14
|
Fan C, Luo Z, Zheng Q, Xu Y, Xu Y, Chen J, Meng Y, Jiang H, Liu K, Xi Y. Cytoglobin augments ferroptosis through autophagic degradation of ferritin in colorectal cancer cells. Mol Cell Biochem 2025; 480:2881-2892. [PMID: 39503803 PMCID: PMC12048458 DOI: 10.1007/s11010-024-05148-0] [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: 09/21/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024]
Abstract
Autophagy has gained importance in the context of ferroptosis. Nevertheless, a deeper understanding of the regulatory mechanism governing autophagy-dependent ferroptosis is necessary. Cytoglobin (CYGB), a member of the globin family, exhibits antifibrotic effects, regulates cellular reactive oxygen species, and stimulates tumor inhibition. Herein, we present further insights into the role of CYGB in ferroptosis regulation. Our investigation confirmed that CYGB impedes cell proliferation and migration. Furthermore, a significant association between CYGB and the lysosomal pathway was suggested based on the RNA sequencing data analysis. Elevated lysosomal signal and colocalization of CYGB with lysosome-associated membrane glycoprotein 1 (LAMP1) were observed. Moreover, upregulated autophagy and augmented ferroptosis induced by RSL3 were confirmed in CYGB-overexpression cells with an obviously increased colocalization of nuclear receptor coactivator 4 (NCOA4) and LC3B. The autophagy inhibitor bafilomycin or chloroquine alleviated autophagy-dependent degradation of ferritin protein under RSL3 treated condition. Additionally, a colocalization of CYGB with the transferrin receptor (TFR) was confirmed. Our results demonstrate an important functional pathway by which CYGB regulates ferroptosis through TFR-binding and autophagic degradation of ferritin, and provide a potential pathway for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Chengjiang Fan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Ziyang Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Institute of Transplantation Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Qingfang Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yuhang Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yao Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jianing Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - You Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Haizhong Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Kaitai Liu
- Department of Radiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315001, China
| | - Yang Xi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
15
|
Xiong X, Huang H, Wang N, Zhou K, Song X. Sirt1 overexpression inhibits chondrocyte ferroptosis via Ftl deacetylation to suppress the development of osteoarthritis. J Bone Miner Metab 2025; 43:203-215. [PMID: 39786573 DOI: 10.1007/s00774-024-01574-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/09/2024] [Indexed: 01/12/2025]
Abstract
INTRODUCTION Osteoarthritis (OA) is a chronic degenerative joint disorder characterized by an imbalance in chondrocyte metabolism. Ferroptosis has been implicated in the pathogenesis of OA. The role of Sirt1, a deacetylase, in mediating deacetylation during ferroptosis in OA chondrocytes remains underexplored. This study aimed to elucidate the mechanisms by which Sirt1 influences chondrocyte ferroptosis in the development of OA. MATERIALS AND METHODS In vitro and in vivo models of OA were established using IL-1β-induced mouse chondrocytes and a destabilization of the medial meniscus (DMM) mouse model, respectively. Ferroptosis was evaluated through measurements of cell viability, lactate dehydrogenase (LDH) release, intracellular levels of Fe2+, glutathione (GSH), malondialdehyde (MDA), lipid reactive oxygen species (ROS), propidium iodide staining, and Western blot analysis. The underlying mechanisms were further investigated using quantitative real-time polymerase chain reaction, Western blotting, immunoprecipitation (IP), co-immunoprecipitation (Co-IP), and glutathione-S-transferase pulldown assays. In vivo validation was performed via Safranin O staining. RESULTS IL-1β induced ferroptosis and increased histone acetylation, effects that were partially reversed by Sirt1 overexpression. Mechanistically, Sirt1 overexpression upregulated ferritin light polypeptide (Ftl) expression by deacetylating Ftl at the K181 residue. Ftl knockdown inhibited the ferroptosis-enhancing effect of Sirt1 overexpression in chondrocytes. In vivo studies showed that Sirt1 overexpression mitigated the progression of OA and reduced ferroptosis in the DMM-induced OA mouse model. CONCLUSION Our findings confirm that Sirt1 overexpression promotes Ftl expression through deacetylation at the K181 site, thereby suppressing chondrocyte ferroptosis and attenuating the progression of OA. These results suggest a potential therapeutic target for OA treatment.
Collapse
Affiliation(s)
- Xiaolong Xiong
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Hui Huang
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ning Wang
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Kai Zhou
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xinghui Song
- Universiti Kebangsaan Malaysia Health Science, UKM, 43600, Bandar Baru Bangi, Selangor, Malaysia.
| |
Collapse
|
16
|
Jin Y, Wu O, Chen Z, Chen L, Zhang K, Chen Q, Tian H, Wang X, Jones M, Kwan KYH, Li YM, Makvandi P, Wang X, Hai X, Zhang J, Wu A. Exploring extracellular vesicles as novel therapeutic agents for intervertebral disc degeneration: delivery, applications, and mechanisms. Stem Cell Res Ther 2025; 16:221. [PMID: 40312404 PMCID: PMC12044939 DOI: 10.1186/s13287-025-04299-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Intervertebral disc degeneration is a multifactorial degenerative disease that poses a significant threat to the health of the elderly population. Current treatments primarily focus on physical therapy, medication, and surgery to alleviate symptoms associated with disc compression but do not address the progression of degeneration. Therefore, this review aimed to explore the potential of extracellular vesicle therapy as a novel preventive strategy to delay degeneration and enhance tissue repair in intervertebral discs. We cover the pathogenic mechanisms underlying intervertebral disc degeneration, including inflammation, apoptosis, pyroptosis, ferroptosis, autophagy dysregulation, and the roles of non-coding RNAs. Subsequently, we discussed the therapeutic potential of extracellular vesicles and their molecular components, such as proteins, RNAs, and lipids, in modulating these pathways to counter intervertebral disc degeneration. We provides a comprehensive review of the significant role of extracellular vesicle cargo in mediating repair mechanisms. It discusses the functional enhancement advantages exhibited by extracellular vesicles under current bioengineering modifications and drug loading. The challenges and future prospects of utilizing extracellular vesicle therapy to treat this degenerative condition are also summarized.
Collapse
Affiliation(s)
- Yuxin Jin
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ouqiang Wu
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhihua Chen
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Linjie Chen
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Kai Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qizhu Chen
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200082, China
| | - Haijun Tian
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinzhou Wang
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Morgan Jones
- Spine Unit, The Royal Orthopaedic Hospital, Bristol Road South, Northfield, Birmingham, B31 2AP, UK
| | - Kenny Yat Hong Kwan
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 5/F Professorial Block, Queen Mary Hospital, 102 Pokfulam Road, Pokfulam, Hong Kong, China
| | - Yan Michael Li
- Department of Neurosurgery, University of Rochester Medical Center, 601 Elm-Wood Ave, Rochester, NY, 14642, USA
| | - Pooyan Makvandi
- University Centre for Research & Development, Chandigarh University, Mohali, 140413, Punjab, India
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Xiangyang Wang
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiang Hai
- Ecological-Environment & Health College (EEHC), Zhejiang A & F University, Hangzhou, 311300, Zhejiang, China.
| | - Jun Zhang
- Department of Orthopedics, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou, China.
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
17
|
Zhong W, Chen R, Zhao J, Zhang Y, He J, Wang H, Zhu F, Fan C, Liu X. SETD7 drives diabetic endothelial dysfunction through FBXO45-mediated GPX4 ubiquitylation. Cardiovasc Diabetol 2025; 24:178. [PMID: 40275362 PMCID: PMC12023459 DOI: 10.1186/s12933-025-02740-6] [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: 01/06/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Vasculopathy is the most prevalent complication of diabetes. Endothelial damage, a primary contributor to hyperglycemic vascular complications, impacts macro- and micro-vasculatures, causing functional impairment of multiple organs. SETD7 was initially identified as a transcriptional activator based on its ability to methylate histone 3 lysine 4. However, its function in the context of diabetic endothelial dysfunction remains poorly understood. This study aims to elucidate the involvement and underlying mechanisms of SETD7 in diabetic endothelial dysfunction. METHODS SETD7 knockout mice were generated to investigate the effects of SETD7 on Streptozotocin (STZ)-induced hyperglycemia and vascular endothelial injury. Endothelial-specific SETD7 interruption adeno-associated virus (AAV) system was utilized to investigate the effects of SETD7 on diabetic vascular endothelial injury in BKS-DB(Lepr) KO/KO (db/db) mice. In vitro manipulation of SETD7 activation or knockdown was conducted to assess its regulation on the lipid peroxidation, oxidative stress, and cell function of primary rat aortic endothelial cells (RAECs) under high glucose conditions. RESULTS Our study revealed that knockout and endothelial deficiency of SETD7 partially restored damaged vascular function and attenuated the inflammatory response caused by high glucose in both STZ-induced and db/db mice. Moreover, SETD7 activation aggravated oxidative stress injury and resulted in profound dysfunction through Glutathione Peroxidase 4 (GPX4)-mediated lipid peroxidation in RAECs. Mechanistically, SETD7 deficiency reduced p53 mono-methylation and blocked FBXO45 transcription, thereby inhibiting the protein degradation of GPX4 and subsequent lipid peroxidation as well as oxidative stress. CONCLUSIONS In summary, our study demonstrates that SETD7-p53-FBXO45-GPX4 is involved in high glucose-induced oxidative stress injury and exacerbated endothelial dysfunction, which offering great significance for mitigating hyperglycemia-induced endothelial damage.
Collapse
MESH Headings
- Animals
- Mice, Knockout
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/pathology
- Oxidative Stress
- Ubiquitination
- Endothelial Cells/enzymology
- Endothelial Cells/pathology
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Histone-Lysine N-Methyltransferase/deficiency
- Male
- Glutathione Peroxidase/metabolism
- Glutathione Peroxidase/genetics
- F-Box Proteins/metabolism
- F-Box Proteins/genetics
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/enzymology
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/physiopathology
- Cells, Cultured
- Signal Transduction
- Mice, Inbred C57BL
- Rats
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/pathology
- Lipid Peroxidation
- Blood Glucose/metabolism
Collapse
Affiliation(s)
- Wen Zhong
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Ruoxue Chen
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Jialin Zhao
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Yuyu Zhang
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Jintao He
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Huibin Wang
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Feng Zhu
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Chunxiang Fan
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China.
| | - Xinhua Liu
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China.
| |
Collapse
|
18
|
Zhang X, Li J, He J, Li Y, Sun D, Zhang W. Glutathion peroxidase 4 (GPX4) and Ribosomal Protein L40 (RPL40) participate in arsenic induced progression of renal cell carcinoma by regulating the NLRP3 mediated classic pyroptosis pathway. Int J Biol Macromol 2025; 310:143129. [PMID: 40239794 DOI: 10.1016/j.ijbiomac.2025.143129] [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: 02/13/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
Epidemiological studies have demonstrated that long-term exposure to high‑arsenic water increases the risk of kidney cancer. Kidney dysfunction can lead to the accumulation of metabolic waste and chronic inflammation, with the latter being a significant factor in tumor development. Therefore, it is crucial to investigate how environmental arsenic exposure affects renal function and inflammation, as well as its potential influence on the progression of renal carcinoma. Additionally, pyroptosis plays an essential role in immune responses and the maintenance of cellular homeostasis. However, the role and mechanisms of pyroptosis in arsenic-induced kidney cancer progression remain unexplored. Our findings indicated that low-dose arsenic exposure reduces pyroptosis and promotes abnormal proliferation of renal tubular epithelial cells, while high-dose exposure enhances pyroptosis and damages renal tissue structure in mouse models. Mechanistically, in vitro studies confirmed that low-dose arsenic exposure promotes the progression of renal cell carcinoma by downregulating NLRP3 and inhibiting pyroptosis, whereas high-dose exposure has the opposite effect. Proteomics analysis identified GPX4 and RPL40 as key proteins mediating pyroptosis induced by low and high doses of arsenic, respectively. Furthermore, GPX4 and RPL40 were shown to regulate the malignant progression of renal cell carcinoma through their effects on NLRP3-mediated pyroptosis. This study reveals that arsenic exposure induces pyroptosis via NLRP3, leading to renal injury and influencing the malignant progression of renal cancer. Notably, GPX4 and RPL40 regulate this progression under low and high-dose arsenic exposure, respectively.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University), Harbin 150081, China; Joint Key Laboratory of Endemic Diseases (Harbin Medical University Guizhou Medical University Xi'an Jiaotong University), Harbin 150081, China
| | - Jinyu Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University), Harbin 150081, China; Joint Key Laboratory of Endemic Diseases (Harbin Medical University Guizhou Medical University Xi'an Jiaotong University), Harbin 150081, China
| | - Jing He
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University), Harbin 150081, China; Joint Key Laboratory of Endemic Diseases (Harbin Medical University Guizhou Medical University Xi'an Jiaotong University), Harbin 150081, China
| | - Yuanyuan Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University), Harbin 150081, China; Joint Key Laboratory of Endemic Diseases (Harbin Medical University Guizhou Medical University Xi'an Jiaotong University), Harbin 150081, China
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University), Harbin 150081, China; Joint Key Laboratory of Endemic Diseases (Harbin Medical University Guizhou Medical University Xi'an Jiaotong University), Harbin 150081, China.
| | - Wei Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University), Harbin 150081, China; Joint Key Laboratory of Endemic Diseases (Harbin Medical University Guizhou Medical University Xi'an Jiaotong University), Harbin 150081, China.
| |
Collapse
|
19
|
Zhang Y, Zhang D, Liao X, Xu Q, Bu L, Zheng J, Shen P, Yang C. Novel insights into the role of ferroptosis in temporomandibular joint osteoarthritis and knee osteoarthritis. Int J Med Sci 2025; 22:2119-2131. [PMID: 40303500 PMCID: PMC12035841 DOI: 10.7150/ijms.107057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by pain, limited movement, and joint stiffness, significantly impacting the quality of life and imposing substantial economic burdens. This review paper delves into the novel insights of ferroptosis, an iron-dependent form of cell death associated with lipid peroxidation, in the context of temporomandibular joint osteoarthritis (TMJ OA) and knee osteoarthritis (KOA). We explore the pathogenic characteristics of OA, including synovitis, chondrocyte death, and extracellular matrix (ECM) degradation, and discuss the limitations of current therapeutic interventions. Emerging evidence suggests a significant relationship between ferroptosis and OA, with iron accumulation and lipid peroxidation observed in osteoarthritic cartilage. This review highlights the role of ferroptosis in chondrocyte malfunction and apoptosis, inflammation, and extracellular matrix breakdown, which are central to OA pathogenesis. We also discuss potential therapeutic targets, such as Transient Receptor Potential Vanilloid 1 (TRPV1), Glutathione Peroxidase 4 (GPX4), and Nuclear Factor Erythroid 2-Related Factor 2 (NRF2), which modulate ferroptosis and OA progression. The paper consolidates studies on ferroptosis in OA, offering a comprehensive understanding of its role and the development of innovative therapies targeting this cell death mechanism to improve treatment outcomes for OA patients.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
- Shanghai Key Laboratory of Orthopedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Dahe Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Xiaoyu Liao
- Department of Rehabilitation Medicine, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215008, China
| | - Qingyu Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Lingtong Bu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Jisi Zheng
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Pei Shen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Chi Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| |
Collapse
|
20
|
Chen H, Xu P, Mao L, Wang Y, Feng Z, Wang Z, Shen C, Xu Y. CCL3 correlates with ferroptosis in intervertebral disc degeneration and its prognostic significance. Sci Rep 2025; 15:12146. [PMID: 40204911 PMCID: PMC11982295 DOI: 10.1038/s41598-025-94989-w] [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: 10/17/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent and debilitating condition associated with low back pain (LBP). Despite its significant impact, effective diagnostic markers for early-stage IVDD remain elusive. Recent research has implicated ferroptosis, a newly recognized form of programmed cell death, in the pathogenesis of IVDD, particularly involving disturbances in iron homeostasis. Additionally, the CC Chemokine Ligand 3 (CCL3) has been linked to macrophage migration and the progression of IVDD, yet its precise diagnostic and prognostic utility remains uncertain. This study aims to elucidate the underlying mechanisms of ferroptosis and the involvement of CCL3 in IVDD, with the objective of establishing their diagnostic and prognostic significance. By uncovering these mechanisms, novel biomarkers and therapeutic targets for the diagnosis and prognosis of IVDD may be identified. Single-cell sequencing data were acquired from the TCGA database, and a range of bioinformatics methods were employed for comprehensive analysis. Furthermore, validation experiments were conducted using in vitro techniques, including the analysis of human tissue samples, co-culture assays with neutralizing antibodies, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blotting. Our findings suggest that CCL3 holds promise as a diagnostic and may was prognostic biomarker for IVDD. Validation experiments demonstrated that CCL3 functions via the pAMPK/AMPK pathway, thereby modulating apoptosis and impacting the progression of IVDD. Our study underscores the diagnostic and prognostic potential of CCL3 in patients with IVDD. Further investigations are warranted to explore therapeutic strategies targeting CCL3, ultimately enhancing the management of IVDD.
Collapse
Affiliation(s)
- He Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, Anhui Province, China
| | - Peng Xu
- Department of Spinal Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Leijing Mao
- Department of Expanded Program on Immunization, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui Province, China
| | - Yicong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zian Feng
- Department of Cardiology, Division of Life Sciences and Medicine, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Zhongxin Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Cailiang Shen
- Department of Spinal Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China.
| | - Yuanhong Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China.
| |
Collapse
|
21
|
Zhou X, Pan Y, Li J, Zhuang R, Tong P, Xia H. Notopterol mitigates osteoarthritis progression and relieves pain in mice by inhibiting PI3K/Akt/GPX4-mediated ferroptosis. Int Immunopharmacol 2025; 151:114323. [PMID: 40020461 DOI: 10.1016/j.intimp.2025.114323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/11/2025] [Accepted: 02/14/2025] [Indexed: 03/03/2025]
Abstract
Ferroptosis-induced lipid peroxidation in chondrocytes exacerbates intra-articular inflammation, oxidative stress, and articular cartilage degradation, accelerating osteoarthritis (OA) progression. Effective anti-inflammatory and antioxidant interventions can alleviate both joint pain and cartilage damage. This study aims to elucidate the therapeutic effects of Notopterol (NP), a bioactive compound extracted from the rhizome of Notopterygium incisum, a traditional Chinese medicine known for its potent anti-inflammatory and antioxidant properties, in treating OA. An in vivo mouse model of OA was established through medial meniscus destabilization (DMM). Intra-articular injections of NP over a 4-week treatment period significantly alleviated pain and gait abnormalities, reduced subchondral osteosclerosis, and attenuated cartilage degradation compared to the untreated DMM group. In vitro, chondrocytes treated with IL-1β to simulate OA conditions exhibited increased viability following NP pretreatment, with concurrent reductions in apoptosis, reactive oxygen species (ROS) accumulation, and chondrocyte catabolic dysfunction, along with enhanced extracellular matrix (ECM) synthesis. Mechanistically, NP exerts its anti-OA effects by inhibiting PI3K/Akt phosphorylation, suppressing ferroptosis, and improving antioxidant defense via upregulation of glutathione (GSH) and glutathione peroxidase 4 (GPX4), thereby preventing lipid peroxidation. In conclusion, NP modulates the PI3K/Akt/GPX4 axis to protect against lipid peroxidation, inhibit ferroptosis, and preserve cartilage integrity, thus delaying OA progression.
Collapse
Affiliation(s)
- Xing Zhou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yu Pan
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China; Quzhou TCM Hospital at the Junction of Four Provinces Affiliated toZhejiang Chinese Medical University, Quzhou, Zhejiang, China; Quzhou Hospital of Traditional Chinese Medicine, Quzhou, Zhejiang, China
| | - Jinlei Li
- Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming City, Yunnan Province, China
| | - Rujie Zhuang
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China; Quzhou TCM Hospital at the Junction of Four Provinces Affiliated toZhejiang Chinese Medical University, Quzhou, Zhejiang, China; Quzhou Hospital of Traditional Chinese Medicine, Quzhou, Zhejiang, China.
| | - Peijian Tong
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China; Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, China.
| | - Hanting Xia
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China; Department of Orthopaedics, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, China; Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, China.
| |
Collapse
|
22
|
Xu Y, Gu X, Li X, Chen Y, Wei Z, Wang J, Liu Y, Ji Y, Cheng Q, Jiang S, Yu J, Li X, Cui W, Ye X. β-Diketone Functionalized Microspheres Chelate Reactive Iron via Metal Coordination for Cartilage Repair. Adv Healthc Mater 2025; 14:e2403933. [PMID: 40045641 DOI: 10.1002/adhm.202403933] [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: 10/10/2024] [Revised: 02/23/2025] [Indexed: 04/18/2025]
Abstract
Excessive intracellular iron accumulation can induce mitochondrial dysfunction, leading to chondrocyte ferroptosis, a key contributor to cartilage damage in osteoarthritis (OA). Here, micelle-microfluidic hydrogel microspheres, featuring keto-enol-thiol bridged nano-sized secondary structures that disintegrate within the intracellular peroxidative environment to reveal β-diketone groups with metal chelation capabilities, are utilized for the in situ removal of reactive iron, thereby facilitating cartilage repair through the restoration of mitochondrial homeostasis. The relevant experiments demonstrate that the microspheres reduce iron influx by downregulating transferrin receptor (TfR1) expression and decrease mitochondrial iron uptake by upregulating mitochondrial outer membrane iron-sulfur cluster protein (CISD1), thus restoring intracellular mitochondrial iron homeostasis. Furthermore, the antioxidant properties of the ketone-thioether segments synergistically mitigate chondrocyte phospholipid peroxidation via Nrf2/SLC7A11/GPX4 axis, inhibiting ferroptosis and slowing OA progression. In summary, this system that in situ sustainably chelates reactive iron via metal coordination exhibits great potential in the minimally invasive treatment of OA and other ferroptosis-mediated diseases.
Collapse
Affiliation(s)
- Yong Xu
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Department of Spinal Surgery, Qinghai University Affiliated Hospital, 29 Tongren Road, Xi Ning, Qinghai, 810006, China
| | - Xin Gu
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Xingchen Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yicheng Chen
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Zhenyuan Wei
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Jielin Wang
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Yi Liu
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Yunhan Ji
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Qian Cheng
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Shuai Jiang
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Jiangming Yu
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Xiaoxiao Li
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - XiaoJian Ye
- Department of Orthopaedics, Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Center for Spinal Minimally Invasive Research, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| |
Collapse
|
23
|
Gao Y, Chen X, Duan JA, Xiao P. A review of pharmacological mechanisms, challenges and prospects of macromolecular glycopeptides. Int J Biol Macromol 2025; 300:140294. [PMID: 39863220 DOI: 10.1016/j.ijbiomac.2025.140294] [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: 06/06/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Macromolecular glycopeptides are natural products derived from various sources, distinguished by their structural diversity, multifaceted biological activities, and low toxicity. These compounds exhibit a wide range of biological functions, such as immunomodulation, antitumor effects, anti-inflammatory properties, antioxidant activity, and more. However, limited understanding of natural glycopeptides has hindered their development and practical application. To promote their advancement and utilization, it is crucial to thoroughly investigate the pharmacological mechanisms of glycopeptides and address the challenges in natural glycopeptide research. This review uniquely focuses on the primary biological activities and potential molecular mechanisms of glycopeptides as reported in recent literature. Moreover, we emphasize the current challenges in glycopeptide research, including extraction and isolation difficulties, purification challenges, structural analysis complexities, elucidation of structure-activity relationships, characterization of biosynthetic pathways, and ensuring bioavailability and stability. The future prospects for glycopeptide research are also explored. We argue that ongoing research into glycopeptides will significantly contribute to drug development and provide more effective therapeutic options and disease treatment alternatives for human health.
Collapse
Affiliation(s)
- Ye Gao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Xiaoyi Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Ping Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
24
|
Shi Y, Zhao Y, Sun SJ, Lan XT, Wu WB, Zhang Z, Chen YX, Yan YY, Xu YP, Li DJ, Fu H, Shen FM. Targeting GPX4 alleviates ferroptosis and retards abdominal aortic aneurysm formation. Biochem Pharmacol 2025; 234:116800. [PMID: 39952331 DOI: 10.1016/j.bcp.2025.116800] [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: 07/23/2024] [Revised: 12/16/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Abdominal aortic aneurysm (AAA) is a potentially fatal cardiovascular disease, closely related to inflammation and loss of vascular smooth muscle cells (VSMCs). Ferroptosis is an iron-dependent cell death associated with peroxidation of lipids. However, the direct role of glutathione peroxidase 4 (GPX4) itself determined ferroptosis in the course of AAA pathogenesis remains unknown. Here, we reported that ferroptosis was triggered in human AAA, elastase- and angiotensin II (Ang II)-induced mouse AAA, and Ang II-incubated VSMCs. Inhibition of ferroptosis via global genetic overexpression of GPX4, a critical anti-ferroptosis molecule, markedly prevented both vascular remodeling and inflammatory response. Mechanistically, GPX4 changed the migration and activation of macrophages/monocytes in AAA tissues in mice. Experiments in vitro demonstrated that overexpression of GPX4 prevented the JAK1/STAT3 signaling activation in VSMCs induced by IL-6, production of pro-inflammatory macrophages. Finally, the role of ferroptosis was confirmed on an Ang II-induced mice AAA model. These results emphasized the significance of ferroptosis in AAA, and provided novel insights that therapy focusing on GPX4 might be a promising strategy for treatment of AAA in the clinic.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Ferroptosis/physiology
- Animals
- Aortic Aneurysm, Abdominal/prevention & control
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors
- Phospholipid Hydroperoxide Glutathione Peroxidase/biosynthesis
- Humans
- Mice
- Mice, Inbred C57BL
- Male
- Angiotensin II/toxicity
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cells, Cultured
Collapse
Affiliation(s)
- Yu Shi
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Zhao
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Si-Jia Sun
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiu-Ting Lan
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Bin Wu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Zhen Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu-Xin Chen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu-Ying Yan
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yu-Ping Xu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Hui Fu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Li Z, Liu J, Ju J, Peng X, Zhao W, Ren J, Jia X, Wang J, Gao F. Cordycepin ameliorates morphine tolerance by inhibiting spinal cord ferroptosis and inflammation via targeting SIRT1. Int J Med Sci 2025; 22:2059-2074. [PMID: 40303502 PMCID: PMC12035829 DOI: 10.7150/ijms.108518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
Morphine tolerance caused by long-term use of morphine is a major medical problem. Neuroinflammation plays an important role in morphine tolerance, and currently no drugs have been found for clinical use to alleviate neuroinflammation during morphine tolerance. Cordycepin is the main active component of fungus cordycepin militaris, has been demonstrated to have anti-oxidative stress and anti-inflammatory properties in various diseases. In this study, we established a rat model of morphine tolerance, examined the effect of cordycepin on the development of morphine tolerance, and evaluated its potential regulatory mechanisms. We found that cordycepin treatment ameliorated the development of morphine tolerance, improved mitochondrial damage associated with ferroptosis, by reducing the levels of reactive oxygen species (ROS), malondialdehyde (MDA) and Fe2+, increasing superoxide dismutase (SOD) and glutathione (GSH) levels, and decreasing the secretion of pro-inflammatory factors (IL-1β, IL-6, and TNF-α). Besides, cordycepin upregulated the expression of SIRT1, SLC7A11 and GPX4. Further research found that the above effects of cordycepin on morphine-tolerant rats were abolished by SIRT1 selective inhibitor EX-527. Thus, these findings indicated that cordycepin could ameliorate the development of morphine tolerance by inhibiting spinal cord ferroptosis and inflammation via targeting SIRT1. Collectively, these results demonstrated the protective effects of cordycepin and highlighted its therapeutic potential as a drug component for morphine tolerance treatment and prevention.
Collapse
Affiliation(s)
- Zheng Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Department of Anesthesiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jie Liu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jie Ju
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaoling Peng
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Wei Zhao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jihao Ren
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaoqian Jia
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jihong Wang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Feng Gao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| |
Collapse
|
26
|
El-Sehrawy AAMA, Rashid TA, Ullah MI, Uthirapathy S, Ganesan S, Singh A, Devi A, Joshi KK, Jasim AS, Kadhim AJ. Cutting edge: ferroptosis in metabolic dysfunction-associated steatotic liver disease (MASLD) pathogenesis and therapy. Funct Integr Genomics 2025; 25:71. [PMID: 40131513 DOI: 10.1007/s10142-025-01579-0] [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: 01/06/2025] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Ferroptosis denotes a distinct form of controlled cell death marked by substantial iron buildup and significant lipid peroxidation, playing a crucial role in several disease processes linked to cell death. Given the liver's essential functions in iron and lipid metabolism and its vulnerability to oxidative damage, more research has investigated the correlation between ferroptosis and numerous hepatic diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD). NAFLD has arisen as a worldwide public health concern due to elevated morbidity and high death rates. The pathogenesis of MASLD remains incompletely elucidated. Recent data suggests that ferroptosis is crucial in the pathophysiology of MASLD; nevertheless, the specific processes by which ferroptosis influences MASLD remain unclear. The present review summarizes the molecular processes of ferroptosis and its intricate regulatory networks, outlines the differing impacts of ferroptosis at different stages of MASLD, and examines possible approaches targeting ferroptosis for the therapy of MASLD, suggesting a novel approach for its management.
Collapse
Affiliation(s)
| | - Teeba Ammar Rashid
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq.
| | - Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Aljouf, Saudi Arabia
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, 248002, Uttarakhand, India
- Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| | - Ahmed Salman Jasim
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 5100, Babylon, Iraq
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| |
Collapse
|
27
|
Zhang J, Li Y, Li Y, Liu H. Unraveling the brain-joint axis: genetic, transcriptomic, and cohort insights from neuroticism to osteoarthritis. Mamm Genome 2025:10.1007/s00335-025-10112-4. [PMID: 40080206 DOI: 10.1007/s00335-025-10112-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/10/2025] [Indexed: 03/15/2025]
Abstract
The causal relationships between neuroticism and osteoarthritis (OA) were inconclusive in observational studies. We conducted bidirectional two-sample Mendelian randomization (MR) and transcriptome-wide association studies to determine the associations and the underlying transcriptomic basis. The summary-level genome-wide association study data for any site OA, knee OA, erosive hand OA, and hip OA were mainly derived from UK Biobank, and neuroticism was derived from CTGlab. We then utilized weighted regression and propensity score matching (PSM) models to investigate the relationship between neuroticism and OA in 11,948 participants of European ancestry from the National Health and Nutrition Examination Survey from 2005 to 2018. Bidirectional two-sample MR studies revealed that feelings of being fed-up, a sense of miserableness, mood swings, and a higher neuroticism score were all linked to an increased risk of OA. These factors were specifically associated with OA at various sites, including the knee. Conversely, there was no evidence to suggest that OA had any influence on traits related to neuroticism. In a comprehensive analysis that accounted for variables such as age, sex, blood lipids, blood glucose, body weight, smoking, alcohol consumption, and physical activity, it was determined that mental fluctuation significantly increased the incidence of self-reported OA (OR 1.37, 95% CI 1.20-1.58, P < 0.001) based on weighted regression. Further confirmation was provided by PSM analysis, which showed that mental fluctuation was associated with a higher incidence of self-reported OA (OR 1.28, 95% CI 1.08-1.52, P = 0.004). Moreover, differentially expressed genes were enriched in several biological processes, including the cell cycle, lipid metabolism, RNA processing, and immuno-inflammatory responses. The results revealed significant genetic and population-based associations, as well as underlying mechanisms, between neuroticism and osteoarthritis, supporting the concept of a brain-joint axis.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yingjie Li
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yongzhen Li
- Department of Pediatric, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Hongwei Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
28
|
Tuerxun H, Zhao Y, Li Y, Liu X, Wen S, Zhao Y. Resveratrol alleviates testicular toxicity induced by anti-PD-1 through regulating the NRF2-SLC7A11-GPX4 pathway. Front Immunol 2025; 16:1529991. [PMID: 40145083 PMCID: PMC11937136 DOI: 10.3389/fimmu.2025.1529991] [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: 11/18/2024] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Background Fertility preservation is a critical concern for reproductive-age cancer survivors, as conventional cytotoxic therapies can cause irreversible damage to the reproductive system, potentially depriving them of the ability to have children in the future. Immune checkpoint inhibitors (ICIs), including anti-programmed cell death protein 1 (anti-PD-1), have become a standard therapeutic approach for various malignancies. However, the impact of ICIs on reproductive function and fertility is not well understood and remains a largely unexplored domain. Resveratrol (RSV), a plant-derived compound, has shown potential as an nuclear factor erythroid 2-related factor 2 (NRF2) agonist to counteract reproductive toxicity induced by various diseases, drugs, and environmental toxins. Methods Male C57BL6/J mice with B16 melanoma were assigned into four groups. RSV and ICI/RSV groups received RSV (40 mg/kg) orally every other day for one month, while controls received the vehicle. ICI and ICI/RSV groups were injected with anti-PD-1 antibody (10 mg/kg) weekly, and controls received IgG2b kappa antibody. Parameters like body and testicular weight, sperm concentration, and western blot for ferroptosis markers were measured. Furthermore, oxidative stress biomarkers, lipid oxidation factors, and gonadal hormone levels were quantified using commercial kits. Results Anti-PD-1 therapy caused male reproductive dysfunction, as evidenced by reduced sperm concentration, altered gonadal hormone levels, and disruption of blood-testis barrier (BTB) integrity. Furthermore, ferroptosis was a key mechanism in anti-PD-1-induced testicular dysfunction, characterized by disrupted iron homeostasis, elevated lipid peroxidation, and suppression of the system Xc-/glutathione peroxidase 4 (GPX4) axis. Additionally, anti-PD-1 therapy diminished antioxidant defenses by inhibiting the NRF2 pathway, thereby increasing the susceptibility to ferroptosis. Crucially, RSV treatment ameliorated anti-PD-1-induced reproductive dysfunction. This was achieved by reducing T cell infiltration, lowering interferon-gamma levels, activating the NRF2 pathway, and maintaining iron and lipid homeostasis. Conclusions Our study demonstrates that anti-PD-1 triggers oxidative stress and ferroptosis in the testis, causing male reproductive dysfunction. RSV may offer protection against testicular toxicity associated with anti-PD-1, particularly through its antioxidant and anti-ferroptosis properties.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuguang Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
29
|
Gong S, Lang S, Jiang X, Li X. Paeonol ameliorates ferroptosis and inflammation in chondrocytes through AMPK/Nrf2/GPX4 pathway. Front Pharmacol 2025; 16:1526623. [PMID: 40124777 PMCID: PMC11925900 DOI: 10.3389/fphar.2025.1526623] [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: 11/12/2024] [Accepted: 01/28/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction Chondrocyte ferroptosis is an important component of the pathogenesis of osteoarthritis. Paeonol, the main pharmacologically active ingredient of the Paeonia suffruticosa Andrews, is a natural radical scavenger with potent biological activities, including antioxidant, anti-inflammatory, and cartilage protection effects. However, the molecular mechanisms underlying its role in regulating chondrocytes ferroptosis remain unclear. Methods To investigate the effect of paeonol on ferroptosis and inflammation of chondrocytes through interleukin-1β (IL-1β), the proliferation activity, lipid peroxidation level, endogenous antioxidant capacity, and mitochondrial membrane potential of chondrocytes were evaluated in detail. Intracellular ferrous ion concentration was detected by FerroOrange fluorescent probe staining. Western blotting and immunofluorescence staining were used to detect biomarker proteins of ferroptosis, inflammation, and AMPK/Nrf2/GPX4 signaling pathway proteins. Results The results showed that paeonol significantly depressed IL-1β-induced ferroptosis and inflammation in chondrocytes. Specifically, paeonol protects cell viability, reduces lipid peroxidation damage, maintains mitochondrial function, and inhibits pro-ferroptosis and pro-inflammation biomarker proteins. In addition, the anti-inflammatory ability of paeonol was partially inhibited after the addition of ferroptosis agonist erastin, suggesting that paeonol protects against inflammatory injury in part by inhibiting ferroptosis. Further studies showed that paeonol activated AMPK phosphorylation and promoted Nrf2 nuclear translocation and Keap1 degradation. Finally, the AMPK-Nrf2-GPX4 signaling pathway was confirmed to be the underlying mechanism of paeonol against ferroptosis by the simultaneous use of the AMPK agonist and Nrf2 inhibitor. Conclusion These results indicate that paeonol significantly inhibits IL-1β-induced ferroptosis and inflammation in chondrocytes, and the underlying mechanism of paeonol against ferroptosis is partly through the AMPK/Nrf2/GPX4 axis.
Collapse
Affiliation(s)
- Shuwei Gong
- Department of Orthopedics, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
- Huzhou Basic and Clinical Translation of Orthopedics Key Laboratory, Huzhou, Zhejiang, China
| | - Shuang Lang
- Department of Traditional Chinese Medicine, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
| | - Xuesheng Jiang
- Department of Orthopedics, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
- Huzhou Basic and Clinical Translation of Orthopedics Key Laboratory, Huzhou, Zhejiang, China
| | - Xiongfeng Li
- Department of Orthopedics, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
- Huzhou Basic and Clinical Translation of Orthopedics Key Laboratory, Huzhou, Zhejiang, China
| |
Collapse
|
30
|
Wang D, Pan Y, Chen W, He D, Qi W, Chen J, Yuan W, Yang Y, Chen D, Wang P, Jin H. Nanodrugs Targeting Key Factors of Ferroptosis Regulation for Enhanced Treatment of Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412817. [PMID: 39840543 PMCID: PMC11923906 DOI: 10.1002/advs.202412817] [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: 10/12/2024] [Revised: 01/03/2025] [Indexed: 01/23/2025]
Abstract
Osteoarthritis (OA) is a globally prevalent degenerative joint disease. Recent studies highlight the role of ferroptosis in OA progression. Targeting ferroptosis regulation presents a promising therapeutic strategy for OA; however, current research primarily focuses on single targets associated with ferroptosis. In this study, a reactive oxygen species (ROS)-responsive nanoparticle is developed by linking deferasirox (DEF) and pterostilbene (PTE) with thioketal and incorporating cerium ions (Ce), creating Ce@D&P nanoparticles (NPs), which offer multitarget regulation of ferroptosis. The characteristics of Ce@D&P NPs are evaluated and their therapeutic effects on IL-1β-stimulated chondrocytes are verified. Results show that Ce@D&P NPs reduce ROS levels, mitigate inflammation, chelate iron to inhibit ferroptosis, and balance extracellular matrix (ECM) metabolism in chondrocytes. Mechanistically, transcriptomics and metabolomics analyses suggest that Ce@D&P NPs exerted their effects by regulating oxidative stress and lipid metabolism in chondrocytes. To better treat destabilization of the medial meniscus (DMM)-induced OA in mice, Ce@D&P NPs via intra-articular injection are delivered. The results show that Ce@D&P NPs alleviate cartilage matrix damage and slow OA progression. Overall, the findings indicate that Ce@D&P NPs represent a promising multitarget drug delivery system, and Ce@D&P NPs may be an effective strategy for OA treatment.
Collapse
Affiliation(s)
- Dong Wang
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| | - Yanli Pan
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| | - Wenzhe Chen
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| | - Du He
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| | - Weihui Qi
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| | - Jiali Chen
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| | - Wenhua Yuan
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| | - Yimin Yang
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| | - Di Chen
- Faculty of Pharmaceutical SciencesShenzhen University of Advanced TechnologyShenzhen518107China
| | - Pinger Wang
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| | - Hongting Jin
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Department of Orthopedic SurgeryHangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhou310000China
| |
Collapse
|
31
|
Wang X, Xu L, Wu Z, Lou L, Xia C, Miao H, Dai J, Fei W, Wang J. Exosomes of stem cells: a potential frontier in the treatment of osteoarthritis. PRECISION CLINICAL MEDICINE 2025; 8:pbae032. [PMID: 39781279 PMCID: PMC11705996 DOI: 10.1093/pcmedi/pbae032] [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: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025] Open
Abstract
The aging population has led to a global issue of osteoarthritis (OA), which not only impacts the quality of life for patients but also poses a significant economic burden on society. While biotherapy offers hope for OA treatment, currently available treatments are unable to delay or prevent the onset or progression of OA. Recent studies have shown that as nanoscale bioactive substances that mediate cell communication, exosomes from stem cell sources have led to some breakthroughs in the treatment of OA and have important clinical significance. This paper summarizes the mechanism and function of stem cell exosomes in delaying OA and looks forward to the development prospects and challenges of exosomes.
Collapse
Affiliation(s)
- Xiaofei Wang
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Lei Xu
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Zhimin Wu
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Linbing Lou
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Cunyi Xia
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Wenyong Fei
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jingcheng Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
32
|
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
|
33
|
Zhang Y, Li J, Liu J, Gao Y, Li K, Zhao X, Liu Y, Wang D, Hu X, Wang Z. Ferroptosis in Osteoarthritis: Towards Novel Therapeutic Strategy. Cell Prolif 2025; 58:e13779. [PMID: 39624950 PMCID: PMC11882765 DOI: 10.1111/cpr.13779] [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: 07/08/2024] [Revised: 10/21/2024] [Accepted: 11/09/2024] [Indexed: 01/06/2025] Open
Abstract
Osteoarthritis (OA) is a chronic, degenerative joint disease primarily characterised by damage to the articular cartilage, synovitis and persistent pain, and has become one of the most common diseases worldwide. In OA cartilage, various forms of cell death have been identified, including apoptosis, necroptosis and autophagic cell death. Ever-growing observations indicate that ferroptosis, a newly-discovered iron-dependent form of regulated cell death, is detrimental to OA occurrence and progression. In this review, we first analyse the pathogenetic mechanisms of OA by which iron overload, inflammatory response and mechanical stress contribute to ferroptosis. We then discuss how ferroptosis exacerbates OA progression, focusing on its impact on chondrocyte viability, synoviocyte populations and extracellular matrix integrity. Finally, we highlight several potential therapeutic strategies targeting ferroptosis that could be explored for the treatment of OA.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Genetics and Cell Biology, School of Basic MedicineQingdao UniversityQingdaoChina
- Department of Reproductive MedicineThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Jing Li
- Department of HematologyRizhao People's HospitalRizhaoChina
| | - Jiane Liu
- Department of Genetics and Cell Biology, School of Basic MedicineQingdao UniversityQingdaoChina
- Department of Reproductive MedicineThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yan Gao
- Department of HematologyThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Kehan Li
- Department of Genetics and Cell Biology, School of Basic MedicineQingdao UniversityQingdaoChina
| | - Xinyu Zhao
- Department of Genetics and Cell Biology, School of Basic MedicineQingdao UniversityQingdaoChina
| | - Yufeng Liu
- Department of Genetics and Cell Biology, School of Basic MedicineQingdao UniversityQingdaoChina
| | - Daijie Wang
- International Joint Laboratory of Medicinal Food R&D and Health Products Creation/Biological Engineering Technology Innovation Center of Shandong ProvinceHeze Branch of Qilu University of Technology (Shandong Academy of Sciences)HezeChina
| | - Xiao Hu
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin Diseases; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsInstitute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical CollegeNanjingChina
| | - Zheng Wang
- Department of Genetics and Cell Biology, School of Basic MedicineQingdao UniversityQingdaoChina
- Department of Reproductive MedicineThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
34
|
Xie Y, Lv Z, Li W, Lin J, Sun W, Guo H, Jin X, Liu Y, Jiang R, Fei Y, Wu R, Shi D. JP4-039 protects chondrocytes from ferroptosis to attenuate osteoarthritis progression by promoting Pink1/Parkin-dependent mitophagy. J Orthop Translat 2025; 51:132-144. [PMID: 40129610 PMCID: PMC11930657 DOI: 10.1016/j.jot.2025.01.001] [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: 07/08/2024] [Revised: 12/05/2024] [Accepted: 01/02/2025] [Indexed: 03/26/2025] Open
Abstract
Background Osteoarthritis (OA) is the most common degenerative joint disease, and its main pathological mechanism is articular cartilage degeneration. The purpose of this study was to investigate the role of mitophagy in the pathogenesis of chondrocyte ferroptosis in OA. Methods The expressions of ferroptosis related proteins (GPX4, FTH1, COX2) and ubiquitin-dependent mitophagy related proteins (PARKIN, PINK1) in the intact and injured areas of OA cartilage were analyzed. Nitro oxide JP4-039, a mitochondrial targeting antioxidant, has bifunctional role of targeting mitochondria. Then we evaluated the potential protective effect of JP4-039 in OA using the destabilization of medial meniscus (DMM)-induced OA model, as well as tert-butyl hydrogen peroxide (TBHP)-treated primary mouse chondrocytes and human cartilage explants. Results The concentrations of iron and lipid peroxidation and the expression of ferroptosis drivers in the damaged areas of human OA cartilages were significantly higher than those in the intact cartilage. Pink1/Parkin-dependent mitophagy decreased in the injured area of human OA cartilage and was negatively correlated with ferroptosis. Then, the toxicity and effectiveness of JP4-039 are tested to determine its working concentration. Next, at the molecular biological level, we found that JP4-039 showed the effect of anti-chondrocyte ferroptosis. Moreover, it was verified on DMM-induced OA model mice, that JP4-039 could delay the progression of OA. Finally, JP4-039 was re-verified in vivo and in vitro to inhibit chondrocyte ferroptosis and delay the progression of OA by promoting Pink1/Parkin-dependent mitophagy. Conclusion JP4-039 inhibits ferroptosis of chondrocytes by promoting Pink1/Parkin-dependent mitophagy and delays OA progression.
Collapse
Affiliation(s)
- Ya Xie
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - Weitong Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - JinTao Lin
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - Wei Sun
- Department of Orthopedic, The Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, 214400, China
| | - Hu Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xiaoyu Jin
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Yuan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Ruiyang Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Clinical College of Xuzhou Medical University, Nanjing, 210008, Jiangsu, China
| | - Yuxiang Fei
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Rui Wu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Clinical College of Xuzhou Medical University, Nanjing, 210008, Jiangsu, China
- State key laboratory of pharmaceutical biotechnology, Nanjing University, Nanjing, 210002, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210002, China
| |
Collapse
|
35
|
Dong S, Li X, Xu G, Chen L, Zhao J. Quercetin attenuates the symptoms of osteoarthritis in vitro and in vivo by suppressing ferroptosis via activation of AMPK/Nrf2/Gpx4 signaling. Mol Med Rep 2025; 31:60. [PMID: 39717946 PMCID: PMC11711930 DOI: 10.3892/mmr.2024.13425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/19/2024] [Indexed: 12/25/2024] Open
Abstract
Osteoarthritis (OA) is a common joint disorder involving the cartilage and other joint tissues. Quercetin (QCT) serves a protective role in the development of OA. However, to the best of our knowledge, the regulatory mechanisms of QCT in the progression of OA have not yet been fully elucidated. In order to mimic a model of OA in vitro, IL‑1β was used to stimulate chondrocytes. Furthermore, an in vivo animal model of OA was induced by anterior cruciate ligament transection (ACLT). 5‑Ethynyl‑2'‑deoxyuridine assays, TUNEL assays, ELISAs, western blotting and immunohistochemical assays were conducted to assess the chondroprotective properties of QCT in the development of OA. The results revealed that 100 µM QCT significantly promoted the proliferation, reduced the apoptosis and inflammation, and inhibited the extracellular matrix (ECM) degradation in IL‑1β‑stimulated chondrocytes. Additionally, QCT attenuated the IL‑1β‑induced ferroptosis of chondrocytes, as demonstrated by the reduced lipid reactive oxygen species and Fe2+ levels. Conversely, the inhibitory effects of QCT on the apoptosis and inflammatory responses were reversed by the activation of ferroptosis by erastin in IL‑1β‑stimulated chondrocytes. Furthermore, QCT significantly elevated the level of phosphorylated (p‑)5' AMP‑activated protein kinase (AMPK) and the levels of two negative regulators of ferroptosis [nuclear factor erythroid 2‑related factor 2 (Nrf2) and glutathione peroxidase 4 (Gpx4)] in IL‑1β‑stimulated chondrocytes. The AMPK inhibitor compound C notably reversed the promoting effects of QCT on phosphorylated‑AMPK, Nrf2 and Gpx4 expression in IL‑1β‑stimulated chondrocytes. Additionally, QCT markedly ameliorated the destruction and degradation of articular cartilage, and elevated the p‑AMPK, Nrf2 and Gpx4 levels in the mouse model of ACLT‑induced OA. Overall, the present study demonstrated that QCT inhibited the development of OA by suppressing ferroptosis via the activation of the AMPK/Nrf2/Gpx4 signaling pathway. These findings provide novel insights into the regulatory mechanisms of QCT for the treatment of patients with OA.
Collapse
Affiliation(s)
- Shiyu Dong
- Department of Orthopedics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, P.R. China
| | - Xiaoliang Li
- Department of Orthopedics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, P.R. China
| | - Genrong Xu
- Department of Orthopedics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, P.R. China
| | - Liming Chen
- Department of Orthopedics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, P.R. China
| | - Jiyang Zhao
- Department of Orthopedics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, P.R. China
| |
Collapse
|
36
|
Liu Y, Song C, Gao S, Zhou D, Lv J, Zhou Y, Wang L, Shi H, Liu F, Xiong Z, Hou Y, Liu Z. Chondrocyte Ferritinophagy as a Molecular Mechanism of Arthritis-A Narrative Review. Cell Biochem Biophys 2025; 83:1021-1033. [PMID: 39306824 DOI: 10.1007/s12013-024-01534-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2024] [Indexed: 03/03/2025]
Abstract
Osteoarthritis (OA) is a prevalent joint disease affecting orthopedic patients. Its incidence is steadily increasing, causing great economic hardship for individuals and society as a whole. OA is connected with risk factors such as genetics, obesity, and joint diseases; yet, its pathophysiology is still largely understood. At present, several cell death pathways govern the initiation and advancement of OA. It has been discovered that the onset and progression of OA are strongly associated with pyroptosis, senescence, apoptosis, ferroptosis, and autophagy. Ferroptosis and autophagy have not been well studied in OA, and elucidating their molecular mechanisms in chondrocytes is important for the diagnosis of OA. For this reason, we aim was reviewed recent national and international developments and provided an initial understanding of the molecular pathways underlying autophagy and ferroptosis in OA. We determined the reference period to be the last five years by searching for the keywords "osteoarthritis, mechanical stress, Pizeo1, ferroptosis, autophagy, ferritin autophagy" in the three databases of PUBMED, Web of Science, Google Scholar. We then screened irrelevant literature by reading the abstracts. Ferroptosis is a type of programmed cell death that is dependent on reactive oxygen species and Fe2+. It is primarily caused by processes linked to amino acid metabolism, lipid peroxidation, and iron metabolism. Furthermore, Piezoelectric mechanically sensitive ion channel assembly 1 (PIEZO1), which is triggered by mechanical stress, has been revealed to be intimately associated with ferroptosis events. It was found that mechanical injury triggers changes in the intracellular environment of articular chondrocytes (e.g., elevated levels of oxidative stress and increased inflammation) through PIEZO1, ultimately leading to iron death in chondrocytes. Therefore, we believe that PIEZO1 is a key initiator protein of iron death in chondrocytes. Widely present in eukaryotic cells, autophagy is a lysosome-dependent, evolutionarily conserved catabolic process that carries misfolded proteins, damaged organelles, and other macromolecules to lysosomes for breakdown and recycling. Throughout OA, autophagy is activated to differing degrees, indicating that autophagy may play a role in the development of OA. According to recent research, autophagy is a major factor in the process that leads cells to ferroptosis. Despite the notion of ferritinophagy being put forth, not much research has been done to clarify the connection between ferroptosis and autophagy in OA.
Collapse
Affiliation(s)
- Yong Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- RuiKang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Chao Song
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- RuiKang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Silong Gao
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Daqian Zhou
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jiale Lv
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yang Zhou
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Liquan Wang
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Houyin Shi
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Fei Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- RuiKang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China.
| | - Zhongwei Xiong
- Luzhou Longmatan District People's Hospital, Luzhou, 646000, Sichuan, China.
| | - Yunqing Hou
- Luzhou Longmatan District People's Hospital, Luzhou, 646000, Sichuan, China.
| | - Zongchao Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Luzhou Longmatan District People's Hospital, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
37
|
Fan M, Chen M, Gao Y, Jiang H, Li Y, Zhu G, Chen S, Xu Y, Chen X. Construction of a novel gene signature linked to ferroptosis in pediatric sepsis. Front Cell Dev Biol 2025; 13:1488904. [PMID: 40070882 PMCID: PMC11893615 DOI: 10.3389/fcell.2025.1488904] [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: 08/30/2024] [Accepted: 01/20/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction Pediatric sepsis is a complex and life-threatening condition characterized by organ failure due to an uncontrolled immune response to infection. Recent studies suggest that ferroptosis, a newly identified form of programmed cell death, may play a role in sepsis progression. However, the specific mechanisms of ferroptosis in pediatric sepsis remain unclear. Methods In this study, we analyzed microarray datasets from pediatric sepsis and healthy blood samples to identify ferroptosis-associated genes. A protein-protein interaction (PPI) network analysis and histological validation were performed to identify key genes. Additionally, immune infiltration analysis was conducted to explore the correlation between immune cells, immune checkpoint-related genes, and key genes. A competing endogenous RNA (ceRNA) network was constructed to investigate potential regulatory mechanisms involving long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and key ferroptosis-related genes. Results We identified 74 genes associated with ferroptosis in pediatric sepsis. Among them, five key genes (MAPK3, MAPK8, PPARG, PTEN, and STAT3) were confirmed through PPI network analysis and histological validation. Immune infiltration analysis revealed significant interactions between immune cells and key genes. The ceRNA network provided insights into the regulatory relationships between lncRNAs, miRNAs, and ferroptosis-related genes. Discussion These findings enhance our understanding of the role of ferroptosis in pediatric sepsis and highlight potential therapeutic targets for future research and clinical interventions.
Collapse
Affiliation(s)
- Mingyuan Fan
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Meiting Chen
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yongqi Gao
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huilin Jiang
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanling Li
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gongxu Zhu
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shengkuan Chen
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiming Xu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiaohui Chen
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
38
|
Song C, Song W, Liu Y, Zhou D, Cai W, Mei Y, Liu F, Jiang F, Chen F, Liu Z. Molecular Mechanisms of Immunoinflammatory Infiltration and Ferroptosis in Arthritis Revealed by a Combination of Bioinformatics and Single-Cell Analysis. J Inflamm Res 2025; 18:2409-2432. [PMID: 39991669 PMCID: PMC11846620 DOI: 10.2147/jir.s503618] [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: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
Background Osteoarthritis (OA) is a widespread chronic inflammatory disease in orthopedics, and its molecular mechanisms are still poorly understood. Objective The purpose of this work was to detect the immunological infiltration of OA and the manner of cell death utilizing bioinformatics and single-cell analysis in order to provide guidelines for clinical therapy and medicine. Methods Ferroptosis -associated genes were sourced from the ferroptosis Database, single-cell and bioinformatic expression profiles were chosen from the Gene Expression Comprehensive Database, and OA gene information was taken from GeneCards. To ascertain the categorization status of OA cells, single-cell analysis was conducted. Protein-protein interaction networks were established by SRING analysis, and functional enrichment was examined in the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) databases. The important proteins of immune-ferroptosis death in OA were elucidated through co-analysis. Last but not least, network pharmacology and molecular docking support the mechanism by which resveratrol controls Ferroptosis in OA. Results The development of OA was found to be tightly related to chondrocytes and immune cells, particularly T and macrophage cells, according to single-cell analysis profile. In patients with OA, immune infiltration also revealed a notable infiltration of T cells, B cells, NK cells, monocytes, and macrophages. The hub genes were shown to be enriched in immunological responses, chemokine-mediated signaling pathways, and inflammatory responses, according to enrichment analysis. The main signaling pathways included autophagy, ferroptosis, the HIF-1 signaling pathway, the PI3K-Akt signaling pathway, and the FoxO signaling pathway. Ferroptosis is a significant cell death mechanism that contributes to the advancement of osteoarthritis. Ferroptosis in chondrocytes is lessened by resveratrol regulation of GPX4, TFRC, and SLC7A11. Conclusion Various immune cell infiltrates, especially T cells and macrophages, play an important role in the progression of OA, and resveratrol ameliorates OA by modulating chondrocyte ferroptosis.
Collapse
Affiliation(s)
- Chao Song
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
- Department of Orthopedics, RuiKang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Weijun Song
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
- Department of Orthopedics, Affiliated Sport Hospital of CDSU, Chengdu, Sichuan Province, People’s Republic of China
| | - Yong Liu
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
| | - Daqian Zhou
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
| | - Weiye Cai
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
| | - Yongliang Mei
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
| | - Fei Liu
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
- Department of Orthopedics, RuiKang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Feng Jiang
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
| | - Feng Chen
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
- Department of Orthopedics, RuiKang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Zongchao Liu
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, People’s Republic of China
- Luzhou Longmatan District People’s Hospital, Luzhou, Sichuan Province, People’s Republic of China
| |
Collapse
|
39
|
Wang X, Liu T, Sheng Y, Qiu C, Zhang Y, Liu Y, Wu C. Exploration and verification of circulating diagnostic biomarkers in osteoarthritis based on machine learning. Front Genet 2025; 16:1513675. [PMID: 40034747 PMCID: PMC11872907 DOI: 10.3389/fgene.2025.1513675] [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: 10/18/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Background Osteoarthritis (OA) is a prevalent chronic joint condition. This study sought to explore potential diagnostic biomarkers for OA and assess their relevance in clinical samples. Methods We searched the GEO database for peripheral blood leukocytes expression profiles of OA patients as a training set to conduct differentially expressed gene (DEG) analysis. Two machine learning algorithms, least absolute shrinkage and selection operator (LASSO) logistic regression and support vector machine-recursive feature elimination (SVM-RFE), were employed to identify candidate biomarkers for OA diagnosis. The performance was assessed using receiver operating characteristic (ROC) curves, and the areas under the curve (AUCs) with 95% confidence interval (CI) were calculated. Furthermore, we gathered clinical peripheral blood samples from healthy donors and OA patients (validation set) to validate our findings. Small interfering RNA and CCK8 proliferation assay were used for experimental verification. Results A total of 31 DEGs were discovered, and the machine learning screening found five DEGs that were considered to be candidate biomarkers. Notably, BIRC2 had a very good discriminatory effect among the five candidate biomarkers, with an AUC of 0.814 (95% CI: 0.697-0.915). In our validation set, results showed that the levels of BIRC2 and SEH1L were remarkably higher in healthy donors than OA patients, consistent with the results of the training set. SEH1L owned the largest AUC of 0.964 (95% CI: 0.855-1.000). BIRC2 also displayed a larger AUC of 0.836 (95% CI: 0.618-1.000) in the training set. Knockdown of these two genes could significantly suppress human chondrocyte proliferation. Conclusion Two novel biomarkers, SEH1L and BIRC2, were indicated to have the capacity to differentiate healthy people from OA patients at the peripheral level. Experiments have shown that knockdown of these two genes could inhibit human chondrocyte proliferation, as verified by cell proliferation assays.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
- Department of Anaesthesia, National Center for Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Tianyi Liu
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yueyang Sheng
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Cheng Qiu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yanzhuo Zhang
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yanqun Liu
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
- Trauma Reconstructive and Plastic Clinical Research Center, Yanbian University Hospital, Jilin, China
| | - Chengai Wu
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Fan S, Wang K, Zhang T, Deng D, Shen J, Zhao B, Fu D, Chen X. Mechanisms and Therapeutic Potential of GPX4 in Pain Modulation. Pain Ther 2025; 14:21-45. [PMID: 39503961 PMCID: PMC11751247 DOI: 10.1007/s40122-024-00673-8] [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: 08/14/2024] [Accepted: 10/04/2024] [Indexed: 01/23/2025] Open
Abstract
Pain, a complex symptom encompassing both sensory and emotional dimensions, constitutes a significant global public health issue. Oxidative stress is a pivotal factor in the complex pathophysiology of pain, with glutathione peroxidase 4 (GPX4) recognized as a crucial antioxidant enzyme involved in both antioxidant defense mechanisms and ferroptosis pathways. This review systematically explores GPX4's functions across various pain models, including neuropathic, inflammatory, low back, and cancer-related pain. Specifically, the focus includes GPX4's physiological roles, antioxidant defense mechanisms, regulation of ferroptosis, involvement in signal transduction pathways, and metabolic regulation. By summarizing current research, we highlight the potential of GPX4-targeted therapies in pain management.
Collapse
Affiliation(s)
- Shiwen Fan
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, 832002, China
| | - Kaixin Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Jiwei Shen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Bowen Zhao
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, 832002, China
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Daan Fu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
41
|
Yang C, Chen X, Liu J, Wang W, Sun L, Xie Y, Chang Q. Identification and Validation of Pivotal Genes in Osteoarthritis Combined with WGCNA Analysis. J Inflamm Res 2025; 18:1459-1470. [PMID: 39906135 PMCID: PMC11792882 DOI: 10.2147/jir.s504717] [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: 11/21/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
Introduction The prevalence of osteoarthritis (OA), the most common chronic joint condition, is increasing due to the aging population and escalating obesity rates, leading to a significant impact on human health and well-being. Thus, analyzing the key targets of OA through bioinformatics can help discover new biomarkers to improve its diagnosis. Methods The microarray and RNA-seq results were screened from the Gene Expression Omnibus (GEO) database. Functional enrichment analyses, protein-protein interaction (PPI) analysis, and weighted gene co-expression network analysis (WGCNA) of the DEGs were performed. RT-qPCR and WB were further performed to verify the hub gene expression in OA rat. Results In this study, 35 key genes were identified through differential expression analysis and weighted gene co-expression network analysis (WGCNA) using the GSE169077 and GSE114007 datasets. Enrichment analysis revealed that these key genes were predominantly enriched in the HIF-1 signaling pathway, ECM-receptor interaction, and FoxO signaling pathway. Through the integration of protein-protein interaction (PPI) analysis, validation in animal models and ROC curve analysis, four pivotal genes (GADD45B, CLDN5, HILPDA and CDKN1B) were finally identified. Conclusion In conclusion, these identified key genes could serve as novel targets for predicting and treating OA, offering fresh insights into its etiology and pathogenesis.
Collapse
Affiliation(s)
- Chengzhuo Yang
- Department of The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xinhua Chen
- Department of The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jin Liu
- Department of The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Wenhao Wang
- Department of The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Lihua Sun
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Youhong Xie
- Department of The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Qing Chang
- Department of The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
42
|
Yu FF, Zuo J, Wang M, Yu SY, Luo KT, Sha TT, Li Q, Dong ZC, Zhou GY, Zhang F, Guo X, Ba Y, Wang YJ. Selenomethionine alleviates T-2 toxin-induced articular chondrocyte ferroptosis via the system Xc -/GSH/GPX4 axis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117569. [PMID: 39700767 DOI: 10.1016/j.ecoenv.2024.117569] [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: 09/20/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
T-2 toxin can induce bone and cartilage development disorder, and oxidative stress plays an important role in it. It is well known that selenomethionine (Se-Met) has antioxidative stress properties and promotes the repair of cartilage lesion, but it remains unclear whether Se-Met can relieve damaged cartilage exposure to T-2 toxin. Here, the oxidative stress and ferroptosis of chondrocytes exposure to T-2 toxin were observed. Mechanistically, T-2 toxin increased ROS, lipid ROS, MDA and Fe2+ contents in chondrocytes, decreased GSH and GPX4 activity, and inhibited the system Xc-/GSH/GPX4 antioxidant axis. In addition, the mitochondria of chondrocytes shrunk and the mitochondrial crest decreased or disappeared. However, Fer-1 (Ferrostatin-1) inhibited ferroptosis induced by T-2 toxin in chondrocytes. The Se-Met alleviated lipid peroxidation, oxidative stress, and damaged mitochondrial in T-2 toxin-infected chondrocytes, enhanced antioxidant enzyme activity, and activated the system Xc-/GSH/GPX4 axis, thereby antagonizing ferroptosis of chondrocytes and alleviating articular cartilage damage. In conclusion, our findings highlight the essentiality of ferroptosis in chondrocyte caused by T-2 toxin, elucidate how Se-Met offers protection against this injury and provide research evidence for the drug treatment target of Kashin-Beck disease.
Collapse
Affiliation(s)
- Fang-Fang Yu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Juan Zuo
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China; Department of Medical Technology, Zhengzhou Shuqing Medical College, Zhengzhou, Henan 450064, PR China.
| | - Miao Wang
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Shui-Yuan Yu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Kang-Ting Luo
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Tong-Tong Sha
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Qian Li
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Zai-Chao Dong
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Guo-Yu Zhou
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Feng Zhang
- Institute of Endemic Diseases, School of Public Health of Health Science Center, Xi'an, Jiaotong University, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an 710061, PR China.
| | - Xiong Guo
- Institute of Endemic Diseases, School of Public Health of Health Science Center, Xi'an, Jiaotong University, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an 710061, PR China.
| | - Yue Ba
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Yan-Jie Wang
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| |
Collapse
|
43
|
Tang J, Liu W, Li Z, Shen C, Zhang L, Wang C, Wang F, Zai Z, Qian X, Hu W, Zhang X, Peng X, Xu Y, Chen F. Inhibition of ASIC1a reduces ferroptosis in rheumatoid arthritis articular chondrocytes via the p53/NRF2/SLC7A11 pathway. FASEB J 2025; 39:e70298. [PMID: 39760183 PMCID: PMC11712546 DOI: 10.1096/fj.202402134rr] [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: 09/09/2024] [Revised: 12/15/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
The activation of acid-sensing ion channel 1a (ASIC1a) in response to extracellular acidification leads to an increase in extracellular calcium influx, thereby exacerbating the degeneration of articular chondrocytes in rheumatoid arthritis (RA). It has been suggested that the inhibition of extracellular calcium influx could potentially impede chondrocyte ferroptosis. The cystine transporter, solute carrier family 7 member 11 (SLC7A11), is recognized as a key regulator of ferroptosis. Recent studies suggest that the tumor suppressor gene p53 facilitates the induction of ferroptosis by suppressing the upregulation of SLC7A11. This process is mediated by the nuclear factor erythroid 2-related factor 2 (NRF2), a key transcription factor integral to the maintenance of cellular redox homeostasis and the regulation of inflammatory responses. This study aims to investigate the role of ASIC1a in the ferroptosis of RA chondrocytes and to determine the involvement of the p53/NRF2/SLC7A11 pathway in its underlying mechanism. In vitro experiments revealed that acidosis induces ferroptosis and reduces the expression of NRF2 and SLC7A11 in chondrocytes. Moreover, acidification significantly increased p53 protein levels in chondrocytes. Pifithrin-α (PFN-α), a p53 inhibitor, mitigated acidosis-induced ferroptosis and restored the diminished expression of NRF2 and SLC7A11. Furthermore, PcTx-1, an ASIC1a inhibitor, inhibited acidification-induced ferroptosis, enhanced the protein levels of SLC7A11 and NRF2, and reduced p53 expression. In vivo experiments demonstrated that the ASIC1a-specific inhibitor PcTx-1 ameliorated histopathological characteristics of ankle joints in collagen-induced arthritis (CIA) mice, decreased p53 expression, and enhanced NRF2 and SLC7A11 expression in chondrocytes. These findings suggest that ASIC1a inhibition may mitigate acidification-induced ferroptosis in articular chondrocytes in RA, potentially via the p53/NRF2/SLC7A11 pathway.
Collapse
Affiliation(s)
- Jie Tang
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Wenqiang Liu
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Zihan Li
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Can Shen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Longbiao Zhang
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Cheng Wang
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Fengshuo Wang
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Zhuoyan Zai
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Xuewen Qian
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Weirong Hu
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Xiaoyue Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Xiaoqing Peng
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Yayun Xu
- Shenzhen Institute of Translational MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's HospitalShenzhenChina
| | - Feihu Chen
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| |
Collapse
|
44
|
Ogawa T, Yokota S, Chen L, Ogawa Y, Nishida Y, Tokuhiro T, Alhasan H, Yutani T, Shimizu T, Takahashi D, Miyazaki T, Endo T, Kadoya K, Terkawi MA, Iwasaki N. Therapeutic Potential of Targeting Ferroptosis in Periprosthetic Osteolysis Induced by Ultra-High-Molecular-Weight Polyethylene Wear Debris. Biomedicines 2025; 13:170. [PMID: 39857757 PMCID: PMC11762349 DOI: 10.3390/biomedicines13010170] [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: 12/24/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Periprosthetic osteolysis is the primary cause of arthroplasty failure in the majority of patients. Mechanistically, wear debris released from the articulating surfaces of a prosthesis initiates local inflammation and several modes of regulated cell death programs, such as ferroptosis, which represents a promising therapeutic target in various chronic inflammatory diseases. Thus, the current study aimed at exploring the therapeutic potential of targeting ferroptosis in a polyethylene-wear-debris-induced osteolysis model. Methods: Inverted cell culture model was used for stimulating the cells with wear debris in vitro, and calvarial osteolysis model was used for evaluating the therapeutic effects of inhibitors in vivo. Results: The immunostaining of periprosthetic bone tissues demonstrated a number of osteocytes expressing ferroptosis markers. Likewise, the expressions of ferroptosis markers were confirmed in polyethylene-wear-debris-stimulated osteocyte-like cells and primary osteoblasts in a direct stimulation model but not in an indirect stimulation model. Furthermore, polyethylene wear debris was implanted onto calvarial bone and mice were treated with the ferroptosis inhibitors DFO and Fer-1. These treatments alleviated the inflammatory and pathological bone resorption induced by the wear debris implantation. Conclusions: Our data broaden the knowledge of the pathogenesis of periprosthetic osteolysis and highlight ferroptosis as a promising therapeutic target.
Collapse
Affiliation(s)
- Takuya Ogawa
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Shunichi Yokota
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Liyile Chen
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Yuki Ogawa
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Yoshio Nishida
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Taiki Tokuhiro
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Hend Alhasan
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Tomoyo Yutani
- R&D Center, Teijin Nakashima Medical Co., Ltd., Okayama 701-1221, Japan;
| | - Tomohiro Shimizu
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Daisuke Takahashi
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Takuji Miyazaki
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Tsutomu Endo
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Ken Kadoya
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Mohamad Alaa Terkawi
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (T.O.); (S.Y.); (L.C.); (Y.O.); (Y.N.); (T.T.); (H.A.); (T.S.); (D.T.); (T.M.); (T.E.); (K.K.); (N.I.)
| |
Collapse
|
45
|
Zhang B, Xiao Y, Su D, Li C, Zhang S, Long J, Weng R, Liu H, Chen Y, Liao Z, Zhu X, Huang J, Chen S, Zhou T, Ma Y, Xu C. M13, an anthraquinone compound isolated from Morinda officinalis alleviates the progression of the osteoarthritis via the regulation of STAT3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156329. [PMID: 39706062 DOI: 10.1016/j.phymed.2024.156329] [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: 08/11/2024] [Revised: 11/19/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is characterized by the progressive deterioration of articular cartilage, leading to joint pain and functional impairment. OA severely impacts quality of life and presents a substantial societal burden. Currently, effective treatment options remain limited. Morinda officinalis (MO), a traditional Chinese herb, is commonly used to treat rheumatoid arthritis and alleviate joint pain. M13, an anthraquinone extracted from MO, has shown significant anti-inflammatory properties, making it a promising candidate for the treatment of OA. However, its role in inhibiting OA progression and the mechanisms involved remain poorly understood. PURPOSE The objective of this study is to examine the impact of M13 on osteoarthritis and uncover the mechanisms. METHODS The effects of M13 on OA were assessed using TNF-α induced chondrocyte models and mice with destabilization of the medial meniscus (DMM). Celecoxib was used as a positive control. We evaluated the expression of factors related to chondrocyte degeneration and inflammation through qRT-PCR, immunoblotting, and immunofluorescence. Chondrocyte viability was measured using CCK-8 assays, EdU staining, and flow cytometry. Molecular docking, molecular dynamics simulations and isothermal titration calorimetry (ITC) were performed to evaluate the binding efficacy of target proteins. Additionally, the therapeutic effects of M13 in OA mice were confirmed through in vivo experiments. RESULTS In primary murine chondrocytes, M13 rescued TNF-α-induced matrix degradation and loss of vitality while suppressing ROS generation. Mechanistically, STAT3 was identified as a target protein of M13, through which M13 mitigated OA by inhibiting the STAT3 signaling pathway. Further in vivo experiments demonstrated that M13 reduced the scores of the Osteoarthritis Research Society International (OARSI), alleviating cartilage impairment. M13 enhanced levels of collagen II and aggrecan in cartilage tissue while decreasing the amounts of cartilage-degrading proteins ADAMTS-5 and MMP13. CONCLUSION This is the first study to validate that M13 mitigates the inflammation and damage in cartilage tissue by blocking the STAT3 signaling pathway. These findings hold promise for enhancing innovative clinical interventions targeting OA.
Collapse
Affiliation(s)
- Baolin Zhang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Ya Xiao
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China
| | - Deying Su
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China
| | - Chuan Li
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China
| | - Shun Zhang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jiahui Long
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China
| | - Ricong Weng
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Hengyu Liu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yingtong Chen
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhiheng Liao
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xu Zhu
- Department of Spine Surgery, the Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830002, China
| | - Junming Huang
- Department of Traditional Chinese Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China
| | - Shuqing Chen
- Department of Traditional Chinese Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China
| | - Taifeng Zhou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.
| | - Yuan Ma
- Department of Spine Surgery, the Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830002, China.
| | - Caixia Xu
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
46
|
Ghosal J, Sinchana VK, Chakrabarty S. Ferroptosis meets microRNAs: A new frontier in anti-cancer therapy. Free Radic Biol Med 2025; 226:266-278. [PMID: 39547521 DOI: 10.1016/j.freeradbiomed.2024.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024]
Abstract
Ferroptosis is an iron-dependent lipid peroxidation-mediated cell death. It is distinct from other types of cellular death and is recognized as a potential target for cancer therapy. This review discusses the mechanisms of ferroptosis, including its induction and inhibition pathways, its role in lipid metabolism, and its connection to various signaling pathways. We also explored the relationship between microRNAs and ferroptosis, highlighting the potential role of miRNAs targeting genes involved in ferroptosis. Role of miRNAs in metabolic reprogramming during carcinogenesis is well documented. We have discussed the role of miRNAs regulating expression of genes involved in iron metabolism, lipid metabolism, and redox metabolism which are associated with regulation of ferroptosis. In conclusion, we addressed various opportunities and challenges identified in ferroptosis research and its clinical implementation stressing the necessity of customized treatment plans based on each patient's unique vulnerability to the disease. Our article provides a complete overview of microRNAs and ferroptosis, with possible implications for cancer therapy.
Collapse
Affiliation(s)
- Joydeep Ghosal
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - V K Sinchana
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
47
|
Wang F, Zhang F, Lin B, Xiao W, Wang X, Wang N. Sarsasapogenin stimulates angiogenesis and osteogenesis coupling to treat estrogen deficiency-induced osteoporosis by activating the GPX4/SLIT3/ROBO1 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156297. [PMID: 39637471 DOI: 10.1016/j.phymed.2024.156297] [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: 08/19/2024] [Revised: 10/26/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Promoting the coupling of osteogenesis and angiogenesis is a crucial strategy for the treatment of postmenopausal osteoporosis (PMOP). Estrogen deficiency induces ferroptosis, which is closely associated with the pathophysiology of PMOP. Sarsasapogenin (SAR) is a natural sapogenin with anti-oxidative effects. However, it is unclear whether SAR has a protective role against the impaired osteogenesis and angiogenesis coupling in PMOP. In this study, we evaluated the efficacy of SAR in estrogen deficiency-induced osteoporosis and explored the underlying mechanisms. METHODS Bone marrow mesenchymal stem cells (BMSCs) and human umbilical vein endothelial cells (HUVECs) were utilized to assess the in vitro effects of SAR on the coupling of osteogenesis and angiogenesis. In vivo experiments involved bilateral ovariectomy (OVX)-induced osteoporosis in mice and glutathione peroxidase 4 (GPX4)-knockout (KO) mice. Mice were orally administered SAR (5 or 10 mg/kg/d) for a duration of 12 weeks. The direct target of SAR was investigated through molecular docking, a cellular thermal shift assay, and surface plasmon resonance. Additionally, RNA sequencing was employed to elucidate the underlying mechanisms. RESULTS SAR treatment improved cell viability and osteogenic differentiation while inhibiting ferroptosis in iron dextran-induced BMSCs. Furthermore, SAR enhanced the production of slit guidance ligand 3 (SLIT3) in these cells, which stimulated angiogenesis by activating its receptor, roundabout human homolog 1 (ROBO1), in HUVECs. An in vitro model of ferroptosis induced by erastin demonstrated that SAR promoted the coupling of osteogenesis and angiogenesis by upregulating the BMSCs-SLIT3/HUVECs-ROBO1 axis. Activation of GPX4 was identified as a contributing factor to the effects of SAR on this coupling. Transfection of GPX4 small interfering RNA (siRNA) in BMSCs negated the impact of SAR on the BMSCs-SLIT3/HUVECs-ROBO1 axis. Additionally, SAR was found to directly interact with GPX4, enhancing protein stability, with an equilibrium dissociation constant of 44.6 μM. Notably, SAR did not increase SLIT3, ROBO1, or indicators of osteogenesis or angiogenesis in GPX4-KO mice. CONCLUSIONS These findings underscore the significance of restoring the GPX4/SLIT3/ROBO1 axis in promoting the coupling of angiogenesis and osteogenesis. SAR mitigates PMOP, in part, by activating the BMSCs-SLIT3/HUVECs-ROBO1 axis, with GPX4 serving as an upstream signaling modulator responsible for SLIT3 production. Our observations provide experimental evidence supporting the clinical application of SAR in the treatment of PMOP.
Collapse
Affiliation(s)
- Fang Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Fanxuan Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Bingfeng Lin
- Tongde Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310007, PR China
| | - Wenlong Xiao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310007, PR China
| | - Xuchen Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Nani Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China; Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China; Tongde Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310007, PR China; School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310007, PR China.
| |
Collapse
|
48
|
Gao ZY, Yan GQ, Su L, He SS, Sheng JE, Wu QC, Huang X, Dai YF. Transcriptomic Analysis and Experimental Verification of Ferroptosis Signature Genes in Osteoarthritis. Int J Rheum Dis 2025; 28:e70083. [PMID: 39835491 DOI: 10.1111/1756-185x.70083] [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: 10/21/2024] [Revised: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
Osteoarthritis is a systemic disease that primarily damages articular cartilage and also affects the synovium, ligaments, and bone tissues. The key mechanisms involved are chondrocyte death and degradation of the extracellular matrix. This study aims to identify differentially expressed genes (DEGs) associated with ferroptosis and investigate their roles in the development of osteoarthritis. We used several methods, such as transcriptomic data analysis, gene enrichment analysis, protein-protein interaction network construction, animal model experiments, and immune cell infiltration analysis. Our examination of the GSE114007 dataset uncovered 2614 DEGs, including 1300 that were upregulated and 1314 that were downregulated. From these, we identified eight ferroptosis-related DEGs (FRGs-DEGs). Functional enrichment analysis showed that these genes are significant for cellular migration and tissue remodeling. They are particularly involved in the HIF-1 and PPAR signaling pathways. Additionally, our immune cell infiltration analysis indicated an increase in M0 and M2 macrophages in osteoarthritis samples, while levels of eosinophils and memory B cells were notably decreased. The receiver operating characteristic curve analysis identified GJA1, TIMP Metallopeptidase Inhibitor 1 (TIMP1), and DPP4 as potential biomarkers for osteoarthritis diagnosis, with area under the curve of 0.91, 0.85, and 0.83, respectively. Moreover, RT-qPCR validation in an osteoarthritis rat model confirmed the upregulation of TIMP1, supporting our bioinformatics results. In summary, our study identifies key FRGs-DEGs and their potential roles in osteoarthritis. This research provides new insights into the disease's molecular mechanisms and suggests innovative therapeutic targets for clinical intervention. Future research should aim to include larger patient cohorts and clinical validation to improve the applicability of these findings.
Collapse
Affiliation(s)
- Zhen-Yu Gao
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Guo-Qing Yan
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Linchong Su
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - San-Shan He
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Jiao-E Sheng
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Qing-Chao Wu
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Xia Huang
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Yu-Fang Dai
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| |
Collapse
|
49
|
Tu S, Zou Y, Yang M, Zhou X, Zheng X, Jiang Y, Wang H, Chen B, Qian Q, Dou X, Bao J, Tian L. Ferroptosis in hepatocellular carcinoma: Mechanisms and therapeutic implications. Biomed Pharmacother 2025; 182:117769. [PMID: 39689515 DOI: 10.1016/j.biopha.2024.117769] [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: 10/12/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024] Open
Abstract
Ferroptosis is a novel form of oxidative cell death, in which highly expressed unsaturated fatty acids on the cell membrane are catalyzed by divalent iron or ester oxygenase to promote liposome peroxidation. This process reduces cellular antioxidant capacity, increases lipid reactive oxygen species, and leads to the accumulation of intracellular ferrous ions, which disrupts intracellular redox homeostasis and ultimately causes oxidative cell death. Studies have shown that ferroptosis induces an immune response that has a dual role in liver disease, ferroptosis also offers a promising strategy for precise cancer therapy. Ferroptosis regulators are beneficial in maintaining cellular homeostasis and tissue health, have shown efficacy in treating diseases of the hepatic system. However, the mechanisms of action and molecular regulatory pathways of ferroptosis in hepatocellular carcinoma (HCC) have not been fully elucidated. Therefore, deciphering the role of ferroptosis and its mechanisms in HCC progression is crucial for treating the disease. In this review, we introduce the morphological features and biochemical functions of ferroptosis, outline the molecular regulatory pathways of ferroptosis, and highlights the therapeutic potential of ferroptosis inhibitors and modulators to target it in HCC.
Collapse
Affiliation(s)
- Shanjie Tu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yuchao Zou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Meiqi Yang
- Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, Liaoning, PR China
| | - Xinlei Zhou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Xu Zheng
- The First Affiliated Hospital of Henan University of TCM, Zhengzhou, Henan, PR China
| | - Yuwei Jiang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Haoran Wang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Buyang Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Qianyu Qian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| | - Jianfeng Bao
- The Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| | - Lulu Tian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
50
|
Gao K, Lv L, Li Z, Wang C, Zhang J, Qiu D, Xue H, Xu Z, Tan G. Natural Products in the Prevention of Degenerative Bone and Joint Diseases: Mechanisms Based on the Regulation of Ferroptosis. Phytother Res 2025; 39:162-188. [PMID: 39513459 DOI: 10.1002/ptr.8366] [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: 07/10/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 11/15/2024]
Abstract
Degenerative bone and joint diseases (DBJDs), characterized by osteoporosis, osteoarthritis, and chronic inflammation of surrounding soft tissues, are systemic conditions primarily affecting the skeletal system. Ferroptosis, a programmed cell death pathway distinct from apoptosis, autophagy, and necroptosis. Accumulating evidence suggests that ferroptosis is intricately linked to the pathogenesis of DBJDs, and targeting its regulation could be beneficial in managing these conditions. Natural products, known for their anti-inflammatory and antioxidant properties, have shown unique advantages in preventing DBJDs, potentially through modulating ferroptosis. This article provides an overview of the latest research on ferroptosis, with a focus on its role in the pathogenesis of DBJDs and the therapeutic potential of natural products targeting this cell death pathway, offering novel insights for the prevention and treatment of DBJDs.
Collapse
Affiliation(s)
- Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Longlong Lv
- Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chenmoji Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiahao Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Daodi Qiu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haipeng Xue
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoqing Tan
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|