1
|
Wang YB, Li ZP, Wang P, Wang RB, Ruan YH, Shi Z, Li HY, Sun JK, Mi Y, Li CJ, Zheng PY, Zhang CJ. Iron dysregulation, ferroptosis, and oxidative stress in diabetic osteoporosis: Mechanisms, bone metabolism disruption, and therapeutic strategies. World J Diabetes 2025; 16:106720. [DOI: 10.4239/wjd.v16.i6.106720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/22/2025] [Accepted: 04/16/2025] [Indexed: 06/13/2025] Open
Abstract
Diabetic osteoporosis (DOP) is a common complication in diabetes, driven by hyperglycemia-induced metabolic disturbances, chronic inflammation, and oxidative stress. This review describes the critical role of iron metabolism dysregulation in DOP pathogenesis, focusing on ferroptosis, a novel iron-dependent cell death pathway characterized by lipid peroxidation and reactive oxygen species (ROS) overproduction. Diabetic conditions exacerbate iron overload, impairing osteoblast function and enhancing osteoclast activity, while triggering ferroptosis in bone cells. Ferroptosis not only accelerates osteoblast apoptosis but also amplifies osteoclast-mediated bone resorption, synergistically promoting bone loss. Furthermore, chronic inflammation and oxidative stress disrupt the balance between bone formation and resorption, with elevated pro-inflammatory cytokines (e.g., tumor necrosis factor-α, interleukin-6) and ROS exacerbating cellular dysfunction. Therapeutic strategies targeting iron metabolism (e.g., deferoxamine) and ferroptosis inhibition (e.g., nuclear factor erythroid 2-related factor 2/heme oxygenase-1 pathway activation, antioxidants like melatonin) demonstrate potential to mitigate DOP progression. Future research should prioritize personalized interventions, clinical trials of iron chelators and antioxidants, and mechanistic studies to refine therapeutic approaches. This review provides a comprehensive framework for understanding DOP pathogenesis and highlights innovative strategies to improve bone health in diabetic patients.
Collapse
Affiliation(s)
- Yao-Bin Wang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhi-Peng Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Peng Wang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Rui-Bo Wang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yu-Hua Ruan
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhen Shi
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hao-Yu Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jin-Ke Sun
- The Third Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yang Mi
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Cheng-Jin Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Peng-Yuan Zheng
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Chang-Jiang Zhang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
2
|
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
|
3
|
Tang Y, Leng J, Luo Y, Luo F. Focusing on ferroptosis in alveolar bone loss during periodontitis: From mechanisms to therapies. Int Immunopharmacol 2025; 156:114683. [PMID: 40252463 DOI: 10.1016/j.intimp.2025.114683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/04/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
Periodontitis is an oral immunoinflammatory disease induced by bacterial infection. During periodontitis, the aggravating destruction of the alveolar bone can result in tooth movement and even tooth loss. Current conventional treatments for periodontitis primarily focus on infection control, but their effectiveness in halting and restoring alveolar bone destruction is limited. To identify additional therapeutic targets, researchers have been dedicated to investigating other pathological mechanisms underlying alveolar bone loss during periodontitis. Recently, findings indicate that ferroptosis plays a role in the development of periodontitis. Ferroptosis is a nonapoptotic type of cell death marked by iron accumulation and lipid peroxidation. Recent investigations have revealed the complex interplay of ferroptosis and inflammation. The positive feedback loop between ferroptosis and inflammation may significantly contribute to the exacerbation of alveolar bone loss. In light of the advancements in research within this field in recent years, this review intends to thoroughly summarize the processes by which ferroptosis aggravates alveolar bone loss during periodontitis, along with relevant ferroptosis-targeted therapeutic agents. By highlighting the latest advancements in this area, we hope this review will inspire researchers to develop novel therapeutic strategies for more effective inflammation control and regeneration of alveolar bone.
Collapse
Affiliation(s)
- Yuting Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Junyan Leng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yankun Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Department of General Dentistry, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Xiao W, Yike W, Gongwen L, Youjia X. Ferroptosis-mediated immune responses in osteoporosis. J Orthop Translat 2025; 52:116-125. [PMID: 40271049 PMCID: PMC12017889 DOI: 10.1016/j.jot.2025.03.011] [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: 12/08/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Osteoporosis is a common systemic metabolic disease, characterized by decreased bone mass and susceptibility to fragility fractures, often associated with aging, menopause, genetics, and immunity. Ferroptosis plays an underestimated yet crucial role in the further impact of immune function changes on osteoporosis. Cell ferroptosis can induce alterations in immune function, subsequently influencing bone metabolism. In this context, this review summarizes several mechanisms of ferroptosis and introduces the latest insights on how ferroptosis regulates immune responses, exploring the interactions between ferroptosis and other mechanisms such as oxidative stress, inflammation, etc. This review elucidates potential treatment strategies for osteoporosis, emphasizing the promising potential of ferroptosis as an emerging target in the treatment of osteoporosis. In conclusion, preparations related to ferroptosis exhibit substantial clinical promise for enhancing bone mass restoration. The translational potential of this article: This review elucidates a nuanced conversation between the immune system and osteoporosis, with ferroptosis serving as the connecting link. These findings underscore the potential of ferroptosis inhibition as a therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Wang Xiao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wang Yike
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liu Gongwen
- Department of Orthopaedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xu Youjia
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
5
|
Khanal V, Carroll M, Carter J, Zhong Y, Chikkamagaluru S, Sato A, Allen R, Wankhade U, Dole N. Lipocalin-2 Regulates Osteocyte Ferroptosis and Osteocyte-Osteoblast Crosstalk via Wnt Signaling to Control Bone Formation. RESEARCH SQUARE 2025:rs.3.rs-6430607. [PMID: 40343339 PMCID: PMC12060985 DOI: 10.21203/rs.3.rs-6430607/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Osteoporosis is a multifactorial disease, and emerging evidence suggests that iron overload contributes to its progression. Here, we identify Lipocalin-2 (LCN2), a cytokine secreted by bone cells with endocrine effects on other tissues, as a local regulator of osteocyte iron metabolism and a mediator of skeletal deterioration. Our findings reveal that LCN2 promotes iron accumulation, mitochondrial dysfunction, and ferroptosis in osteocytes in a process dependent on LCN2 receptor SLC22A17. Genetic ablation of Lcn2 (Dmp1-Cre; Lcn2 fl/fl ) in osteocytes mitigates their ferroptotic vulnerability by preserving mitochondrial integrity and limiting iron overload. Remarkably, LCN2 deletion enhances osteocyte dendricity and lacunocanalicular network, supporting their function in bone remodeling. Mechanistically, we demonstrate that Lcn2 ablation in osteocytes decreases DKK1 and SOST expression in bone, leading to increased Wnt/β-catenin signaling and osteoblast-driven bone formation. Using in vitro and in vivo approaches, we establish the LCN2-SLC22A17 axis as a key pathway linking iron homeostasis, osteocyte dysfunction, and skeletal remodeling. These findings provide insight into a previously unrecognized mechanism underlying iron-driven bone loss and suggest that targeting LCN2 could offer therapeutic potential for osteoporosis.
Collapse
Affiliation(s)
| | | | | | - Ying Zhong
- University of Arkansas for Medical Sciences
| | | | - Amy Sato
- University of Arkansas for Medical Sciences
| | - Ryan Allen
- University of Arkansas for Medical Sciences
| | | | - Neha Dole
- University of Arkansas for Medical Sciences
| |
Collapse
|
6
|
Ma RX, Lin BH, Feng SX, Bu YT, Chen ZH, Huang YX, Li EL, Weng SJ, Yang L. Evaluation of proanthocyanidins in treating Type 2 diabetic osteoporosis via SIRT6/Nrf2/GPX4 pathways. FASEB J 2025; 39:e70487. [PMID: 40178920 DOI: 10.1096/fj.202403032r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/17/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025]
Abstract
This study investigates the therapeutic potential of proanthocyanidins (PAC) in addressing Type 2 diabetic osteoporosis (T2DOP) by activating the SIRT6/Nrf2/GPX4 signaling pathways. T2DOP is characterized by compromised bone structure and heightened oxidative stress, where ferroptosis plays a pivotal role. Utilizing a T2DOP mouse model and MC3T3-E1 cells under high glucose conditions, we evaluated the impact of PAC on bone health and iron homeostasis. Our results, obtained through micro-CT, histological staining, Western blot, and immunofluorescence analyses, revealed reductions in bone density and decreased GPX4 expression in T2DOP conditions, indicating ferroptosis and oxidative stress. However, PAC treatment improved trabecular bone structure, reduced bone marrow adipocytes, decreased oxidative stress, and enhanced expression of key osteogenic proteins. These findings highlight PAC's potential in mitigating T2DOP through the SIRT6/Nrf2/GPX4 pathways, offering promising therapeutic insights for managing diabetic osteoporosis.
Collapse
Affiliation(s)
- Run-Xun Ma
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Bing-Hao Lin
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Si-Xiang Feng
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yi-Tian Bu
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Zi-Hao Chen
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yi-Xun Huang
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - En-Li Li
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - She-Ji Weng
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Lei Yang
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| |
Collapse
|
7
|
Gu H, Yu W, Feng P, Zeng C, Cao Q, Chen F, Wang Z, Shen H, Wu Y, Wang S. Circular RNA circSTX12 regulates osteo-adipogenic balance and proliferation of BMSCs in senile osteoporosis. Cell Mol Life Sci 2025; 82:149. [PMID: 40192802 PMCID: PMC11977094 DOI: 10.1007/s00018-025-05684-y] [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: 06/20/2024] [Revised: 01/03/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Increased adipogenic differentiation and decreased osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) along with slow self-renewal are pivotal causes for decreased bone formation in senile osteoporosis. Circular RNAs (circRNAs) play important roles in cell proliferation and differentiation, and are closely related to osteoporosis. Whether circRNAs orchestrate the adipo-osteogenic balance and the proliferation of BMSCs in osteoporosis remains unclear. We found in this study that circSTX12 was abnormally upregulated in bone sections from osteoporosis patients and in BMSCs from aged mice, as well as in later-generation human BMSCs in culture. Knockdown of circSTX12 in BMSCs resulted in enhanced osteogenesis, decreased adipogenesis, and increased proliferation capacity; circSTX12 overexpression had the opposite effect. RNA pull-down and mass spectrometry revealed the interactions between circSTX12 with CBL and LMO7. At the molecular level, circSTX12 regulated cell fate in BMSCs by competitively binding to CBL, reducing the ubiquitination-mediated degradation of MST1 and thereby activating the Hippo pathway, a key regulator of adipo-osteogenic balance. Knockdown of circSTX12 promoted the nuclear localization of YAP. In addition, our findings suggest that LMO7 mediates circSTX12-induced BMSCs proliferation by regulating the transcription of CCNA2, CCNH, and CCND1. In vivo, injection of antisense oligonucleotides (ASOs) to knockdown circSTX12 promoted bone formation in aged mice. Our results provide evidence for circSTX12 as a regulator of adipo-osteogenic differentiation and proliferation of BMSCs through binding to CBL and LMO7, respectively. Targeting circSTX12 may be a novel approach for osteoporosis treatment.
Collapse
Affiliation(s)
- Huimin Gu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Wenhui Yu
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Pei Feng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Qian Cao
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Fenglei Chen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| | - Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| |
Collapse
|
8
|
Li P, Xu TY, Yu AX, Liang JL, Zhou YS, Sun HZ, Dai YL, Liu J, Yu P. The Role of Ferroptosis in Osteoporosis and Advances in Chinese Herbal Interventions. BIOLOGY 2025; 14:367. [PMID: 40282232 PMCID: PMC12025301 DOI: 10.3390/biology14040367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
OP, a systemic bone disorder marked by reduced bone mass and heightened fracture risk, poses a significant global health burden, particularly among aging populations. Current treatments, including bisphosphonates and calcium supplementation, are limited by adverse effects and incomplete efficacy. Emerging research highlights ferroptosis-an iron-dependent cell death driven by lipid peroxidation-as a critical contributor to OP pathogenesis, characterized by dysregulated iron metabolism, oxidative stress, and lipid peroxide accumulation, which disrupt bone remodeling by impairing osteoblast function and enhancing osteoclast activity. This review elucidates the mechanistic interplay between ferroptosis and OP subtypes (diabetic osteoporosis (DOP), glucocorticoid-induced (GIOP), and postmenopausal osteoporosis (PMOP)) and evaluates the efficacy of Chinese herbal interventions in mitigating ferroptosis-driven bone loss. Key findings reveal that excess iron exacerbates lipid peroxidation via the Fenton reaction, while glutathione peroxidase 4 (GPX4) inactivation and system Xc- inhibition amplify oxidative damage. In DIOP, hyperglycemia-induced ROS and advanced glycation end products suppress osteogenesis, countered by melatonin and naringenin via nuclear factor -related factor 2 (Nrf2)/GPX4 activation. GIOP involves dexamethasone-mediated GPX4 downregulation, mitigated by exosomes and melatonin through phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling. PMOP driven by estrogen deficiency-induced iron overload is alleviated by aconitine and icariin (ICA) via nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways. Chinese herbs, including active compounds (quercetin, gastrodin, ICA, etc.) and formulations (Bugu Shengsui Capsule, Erxian Decoction (EXD), etc.), regulate iron metabolism, enhance antioxidant defenses (Nrf2/heme oxygenase 1(HO-1)), and inhibit lipid peroxidation, effectively restoring bone homeostasis. These findings underscore ferroptosis as a pivotal mechanism in OP progression and highlight the therapeutic promise of Chinese herbs in bridging traditional medicine with modern mechanistic insights. Future research should prioritize elucidating precise molecular targets, optimizing formulations, and validating clinical efficacy to address current therapeutic gaps.
Collapse
Affiliation(s)
- Pan Li
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin 130117, China; (P.L.); (A.-X.Y.); (J.-L.L.); (H.-Z.S.)
| | - Tian-Yang Xu
- Innovation Practice Center, Changchun University of Chinese Medicine, Jilin 130117, China;
| | - Ao-Xue Yu
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin 130117, China; (P.L.); (A.-X.Y.); (J.-L.L.); (H.-Z.S.)
| | - Jing-Ling Liang
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin 130117, China; (P.L.); (A.-X.Y.); (J.-L.L.); (H.-Z.S.)
| | - Ya-Shuang Zhou
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin 130117, China; (P.L.); (A.-X.Y.); (J.-L.L.); (H.-Z.S.)
| | - Huai-Zhu Sun
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin 130117, China; (P.L.); (A.-X.Y.); (J.-L.L.); (H.-Z.S.)
| | - Yu-Lin Dai
- Ginseng Scientific Research Institute, Jilin 130117, China;
| | - Jia Liu
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin 130117, China; (P.L.); (A.-X.Y.); (J.-L.L.); (H.-Z.S.)
| | - Peng Yu
- Innovation and Entrepreneurship College, Changchun University of Chinese Medicine, Jilin 130117, China
| |
Collapse
|
9
|
Yuan Z, Chen Y, Xin Y, Zhang Y, Dong Z, Wang J, Wang X, Yang G, Li S. Key role of the CSE/transsulfuration pathway in macrophage phenotypic change under iron overload. J Trace Elem Med Biol 2025; 88:127611. [PMID: 39914135 DOI: 10.1016/j.jtemb.2025.127611] [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: 08/20/2024] [Revised: 11/22/2024] [Accepted: 01/31/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND Iron homeostasis has a significant impact on the phenotypic transformation of macrophages and is implicated in various diseases. In this study, we evaluated the effect of cystathionine-gamma-lyase (CSE)/transsulfuration pathway in iron-overload induced macrophage phenotype change. METHODS The biochemical parameters, such as qRT-PCR, western blot, fluorescence staining, were assessed both in vitro and in vivo. RESULTS Iron overload disrupts iron metabolism and alters the expression of genes involved in iron transport, resulting in the polarization of macrophages towards the M1 phenotype and an alternating activation state of M2. Meanwhile, excessive iron led to an increase in lipid peroxidation levels and disrupted cysteine metabolism. By utilizing erastin to inhibit SLC7A11 activity and block exogenous cysteine uptake, we were able to observe the exacerbation of the proinflammatory state in macrophages under conditions of cysteine deprivation. The CSE/transsulfuration pathway, serves as the primary route for endogenous cysteine synthesis. In the presence of iron overload, the expression of CSE was upregulated and further enhanced by cysteine deprivation. Deletion of CSE in CSE-knockout mice exacerbated the inflammatory transition of iron-overloaded macrophages by impacting cysteine metabolism and ferritinophagy. CONCLUSION The CSE/transsulfuration pathway regulated macrophage phenotype change under iron-overload, which may offer novel insights into potential therapeutic strategies for iron overload-related disorders.
Collapse
Affiliation(s)
- Zhaoji Yuan
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong 250062, China
| | - Yuxuan Chen
- Department of Cell Biology, Shandong University, Jinan, Shandong 250012, China
| | - Yijun Xin
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong 250062, China
| | - Yong Zhang
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Zihao Dong
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Jianxu Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xiangdong Wang
- Department of Cell Biology, Shandong University, Jinan, Shandong 250012, China
| | - Guang Yang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Siying Li
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong 250062, China.
| |
Collapse
|
10
|
Hong R, Chen B, Wu H, Ding J. Crocin facilitates osteogenesis and angiogenesis by moderating oxidative stress and ferroptosis via Nrf2/GPX4 pathway. Tissue Cell 2025; 93:102675. [PMID: 39874918 DOI: 10.1016/j.tice.2024.102675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/11/2024] [Accepted: 12/06/2024] [Indexed: 01/30/2025]
Abstract
Bone formation is a complex multi-factor process of bone defect healing. Oxidative stress (OS) is predisposed to induce regulatory cell death (RCD), such as ferroptosis. At present, the antioxidant effects of Crocin on erastin induced oxidative damage were studied. The activity of bone marrow mesenchymal stem cells (BMSCs) and human umbilical vein endothelial cells (HUVECs) was detected by CCK-8 and EdU staining. The production of reactive oxygen species (ROS), MDA, SOD and GSH were evaluated. Western blotting assay was used to detect ferroptosis-related proteins. The osteogenic function of BMSCs was determined by alkaline phosphatase (ALP) activity, ALP staining and alizarin red S (ARS) staining. Western blotting and RT-PCR assays were used to detect the expression of osteogenic proteins and genes. Angiogenesis of HUVECs was evaluated by tube formation, RT-PCR, scratch test and Transwell assay. The results showed that Crocin can promote the osteogenic function of BMSCs and angiogenesis of HUVECs. In addition, Crocin protects cells from erastin-induced oxidative injury and inhibits ferroptosis via the Nrf2/GPX4 pathway. These findings suggest that Crocin can promote bone defect healing by regulating OS and inhibiting ferroptosis through the Nrf2/GPX4 pathway.
Collapse
Affiliation(s)
- Ruilong Hong
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Jiangsu 221009, China
| | - Bo Chen
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Jiangsu 221009, China
| | - Hao Wu
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Jiangsu 221009, China
| | - Junwen Ding
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Jiangsu 221009, China.
| |
Collapse
|
11
|
Zhao ZM, Ding JM, Li Y, Wang DC, Kuang MJ. Human umbilical cord mesenchymal stem cell-derived exosomes promote osteogenesis in glucocorticoid-induced osteoporosis through PI3K/AKT signaling pathway-mediated ferroptosis inhibition. Stem Cells Transl Med 2025; 14:szae096. [PMID: 40257841 PMCID: PMC12010878 DOI: 10.1093/stcltm/szae096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 11/30/2024] [Indexed: 04/23/2025] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP), the most common cause of secondary osteoporosis, is characterized by significant bone loss, decreased bone quality, and increased fracture risk. The current treatments for GIOP have several drawbacks. Exosomes are vital for cellular processes. However, very few studies have focused on using human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-EXOs) for GIOP treatment. In vitro and in vivo dexamethasone was used to evaluate the therapeutic effects of hUCMSC-EXOs on GIOP. CCK-8 and EdU assays were used to evaluate cell viability and proliferation, respectively. We conducted an alkaline phosphatase activity assay, alizarin red staining, Western blotting, and real-time PCR to detect the effect on osteogenesis. TMT-labeled quantitative proteomic and bioinformatic analyses were performed. Furthermore, we performed Western blotting, immunofluorescence, reactive oxygen species assays, and lipid peroxidation assays to investigate the regulatory mechanism by which hUCMSC-EXOs affect cell proliferation and osteogenic differentiation. The in vivo effects of hUCMSC-EXOs were evaluated using micro-CT, hematoxylin, and eosin staining, and immunohistochemical staining. We found that hUCMSC-EXOs reversed the inhibitory effects of glucocorticoids on human bone marrow stromal cell (hBMSC) proliferation and osteogenic differentiation and demonstrated that hUCMSC-EXOs reversed GIOP via the PI3K/AKT signaling pathway, inhibiting lipid peroxidation in vitro and in vivo. HUCMSC-EXOs promote hBMSC osteogenesis through the PI3K/AKT signaling pathway, inhibit ferroptosis, and have therapeutic potential for GIOP in mice.
Collapse
Affiliation(s)
- Zhi-Meng Zhao
- Department of Orthopedics, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, People’s Republic of China
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People’s Republic of China
| | - Jia-Ming Ding
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People’s Republic of China
| | - Yu Li
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People’s Republic of China
| | - Da-Chuan Wang
- Department of Spine Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, People’s Republic of China
| | - Ming-Jie Kuang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People’s Republic of China
| |
Collapse
|
12
|
Chen X, Wang J, Zhen C, Zhang G, Yang Z, Xu Y, Shang P. Hepcidin knockout exacerbates hindlimb unloading-induced bone loss in mice through inhibiting osteoblastic differentiation. BMC Musculoskelet Disord 2025; 26:276. [PMID: 40102891 PMCID: PMC11917043 DOI: 10.1186/s12891-025-08515-0] [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: 12/16/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND An oligopeptide hepcidin is encoded by the human HAMP gene (Hamp in mice). Its deficiency can result in iron overload, while excess may lead to iron deficiency. Hepcidin knockout mice exhibited iron accumulation in multiple tissues, accompanied by degeneration of bone microarchitecture and reduced biomechanical properties. Astronauts who are exposed to weightlessness during prolonged spaceflight experience bone loss. After space missions, an interrelation exists between iron stores and bone mineral density (BMD). Bone loss in mice due to unloading is linked to iron excess and involves hepcidin. The potential role of hepcidin in unloading-induced bone loss remains unclear. METHODS Our study conducted relevant experiments using hepcidin knockout mice and their primary osteoblasts as the research subjects. We used the hindlimb unloading (HLU) model and the random positioning machine (RPM) system to simulate weightlessness in vivo and in vitro. RESULTS HLU mice exhibited reduced hepcidin levels in the serum and liver. Hepcidin knockout further diminished BMD and bone mineral content (BMC) in the femurs of HLU mice. Similarly, the bone volume fraction (BV/TV) and connectivity density (Conn.Dn) followed this downward trend, whereas trabecular separation (Tb.Sp) showed an inverse pattern. Moreover, hepcidin knockout decreased the ultimate load and elastic modulus in the tibias of HLU mice. Hepcidin knockout decreased PINP levels in the serum, a commonly used marker for bone formation, alongside elevated iron levels in the serum, liver, and bone of HLU mice. We also found higher serum MDA and SOD levels in these mice. In vitro, experimental data indicated that hepcidin knockout suppresses the osteoblastic differentiation capacity under RPM conditions. Additionally, this condition upregulates SOST protein levels and downregulates LRP6 and β-catenin protein levels in osteoblasts. CONCLUSION Hepcidin knockout exacerbates bone loss in HLU mice, most likely due to reduced osteoblastic activity.
Collapse
Affiliation(s)
- Xin Chen
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
- Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen, 518057, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jianping Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
- Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen, 518057, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Chenxiao Zhen
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
- Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen, 518057, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Gejing Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
- Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen, 518057, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zhouqi Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
- Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen, 518057, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Youjia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Peng Shang
- Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen, 518057, China.
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
13
|
Huang Y, Zhang J, Zhu Y, Zhao R, Xie Z, Qu X, Duan Y, Li N, Tang D, Luo X. BMP9 alleviates iron accumulation-induced osteoporosis via the USP10/FOXO1/GPX4 axis. J Adv Res 2025:S2090-1232(25)00153-5. [PMID: 40068762 DOI: 10.1016/j.jare.2025.03.012] [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: 01/04/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
INTRODUCTION Ferroptosis induced by iron accumulation can disrupt the physiological functions of bone marrow mesenchymal stem cells (BMSCs). BMP9 is an effective osteogenic factor. However, the role of BMP9 and its molecular mechanisms in osteoporosis induced by iron accumulation remain unclear. OBJECTIVES This study aims to explore the role and mechanism of BMP9 in alleviating iron accumulation induced osteoporosis. METHODS Clinical samples were collected to analyze the relationship between iron accumulation and osteoporosis. The effect of BMP9 on lipid peroxidation levels in BMSCs under iron accumulation conditions was assessed using C11-BODIPY staining, MitoSOX staining, MDA and SOD activity measurement. The osteogenic capacity of BMP9 in BMSCs under iron accumulation conditions was evaluated by measuring ALP activity and calcium nodule formation. The mechanisms of BMP9 in regulating BMSCs under iron accumulation conditions were explored through experiments including cycloheximide treatment, RT-PCR, Western blot, GST pull-down, ChIP, and CO-IP. RESULTS It was observed in human samples that serum ferritin levels were negatively correlated with the bone mineral density of the lumbar spine and femoral neck. Meanwhile, ferroptosis is considered a key factor affecting bone health. Further research indicated that BMP9 could inhibit ferroptosis in cells and animal models with iron accumulation, while also improving oxidative stress and osteogenic capacity. In-depth investigation of its mechanism reveals that BMP9 promotes the expression of USP10, which removes the K48-linked ubiquitin chains on FOXO1, inhibiting its excessive ubiquitination in the cytoplasm. This stabilization allows FOXO1 to accumulate in the cytoplasm and eventually re-enter the nucleus. This process activated the expression of the key inhibitor of cell death, GPX4, enhancing the cell's antioxidant response, reducing ferroptosis-induced damage to BMSCs, and promoting their osteogenic differentiation. CONCLUSION This study reveals that BMP9 inhibits ferroptosis through the USP10/FOXO1/GPX4 axis, providing a new therapeutic strategy for osteoporosis caused by iron accumulation.
Collapse
Affiliation(s)
- Yanran Huang
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jun Zhang
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yafei Zhu
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Runhan Zhao
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhou Xie
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiao Qu
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yingtao Duan
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ningdao Li
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Dagang Tang
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Xiaoji Luo
- Department of Orthopaedic Surgery, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine/Orthopaedic Research Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing 400060, China; Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| |
Collapse
|
14
|
Wang D, Shen J, Wang Y, Cui H, Li Y, Zhou L, Li G, Wang Q, Feng X, Qin M, Dong B, Yang P, Li Y, Ma X, Ma J. Mechanisms of Ferroptosis in bone disease: A new target for osteoporosis treatment. Cell Signal 2025; 127:111598. [PMID: 39788305 DOI: 10.1016/j.cellsig.2025.111598] [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: 11/15/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Osteoporosis (OP) is a common disease in the elderly, characterized by decreased bone strength, reduced bone density, and increased fracture risk. There are two clinical types of osteoporosis: primary osteoporosis and secondary osteoporosis. The most common form is postmenopausal osteoporosis, which is caused by decreased estrogen production after menopause. Secondary osteoporosis, on the other hand, occurs when certain medications, diabetes, or nutritional deficiencies lead to a decrease in bone density. Ferroptosis, a new iron-dependent programmed cell death process, is critical in regulating the development of osteoporosis, but the underlying molecular mechanisms are complex. In the pathologic process of osteoporosis, several studies have found that ferroptosis may occur in osteocytes, osteoblasts, and osteoclasts, cell types closely related to bone metabolism. The imbalance of iron homeostasis in osteoblasts and excessive iron accumulation can promote lipid peroxidation through the Fenton reaction, which induces ferroptosis in osteoblasts and affects their role in regulating bone metabolism. Ferroptosis in osteoblasts inhibits bone formation and reduces the amount of new bone production. Osteoclast-associated ferroptosis abnormalities, on the other hand, may alter the homeostasis of bone resorption. In this paper, we start from the molecular mechanism of ferroptosis, and introduce the ways in which ferroptosis affects the physiological and pathological processes of the body. After that, the effects of ferroptosis on osteoblasts and osteoclasts will be discussed separately to elucidate the molecular mechanism between ferroptosis and osteoporosis, which will provide a new breakthrough for the prevention and treatment of osteoporosis and a more effective and better idea for the treatment strategy of osteoporosis.
Collapse
Affiliation(s)
- Dong Wang
- College of Integrative Medicine of Tianjin University of traditional Chinese Medicine, Tianjin 301617,China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin 301617, China
| | - Jiahui Shen
- College of Integrative Medicine of Tianjin University of traditional Chinese Medicine, Tianjin 301617,China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin 301617, China
| | - Yan Wang
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Hongwei Cui
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Yanxin Li
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Liyun Zhou
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Guang Li
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Qiyu Wang
- College of Integrative Medicine of Tianjin University of traditional Chinese Medicine, Tianjin 301617,China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaotian Feng
- College of Integrative Medicine of Tianjin University of traditional Chinese Medicine, Tianjin 301617,China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin 301617, China
| | - Mengran Qin
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Benchao Dong
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Peichuan Yang
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Yan Li
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Xinlong Ma
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Jianxiong Ma
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China.
| |
Collapse
|
15
|
Zhang J, Ma H, Yang Y, Liu L, Luo D, Yu D, Chen T. Iron-lead mixed exposure causes bone damage in mice: A multi-omics analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117967. [PMID: 40037083 DOI: 10.1016/j.ecoenv.2025.117967] [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: 09/04/2024] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 03/06/2025]
Abstract
Excessive intake of essential and toxic metals affects the pathological process of osteoporosis. At present, the effects of single forms of iron (Fe), lead (Pb) and other metals on bone injury have been widely studied. However, these metal elements usually do not exist in the environment in a separate form. They are ingested in various ways and are often found together in the human body. However, the mechanism of bone damage caused by Fe and Pb mixed exposure is still unclear at this stage. At present, the combined analysis of multi-omics is the conventional method to explore the molecular mechanism behind the disease. Therefore, we attempted to combine proteomics and metabolomics to explain the mechanism of bone damage caused by mixed Fe and Pb exposure. Differential proteins and metabolites were found to be predominantly enriched in the JAK-STAT signalling pathway, inflammatory bowel disease (IBD), and osteoclast differentiation. Combined analysis showed that Fpr2, Lifr, Lisofylline, 7-Ketocholesterol, LacCer (d18: 1/14:0) and other substances may be involved in the process of bone injury mediated by mixed metal exposure. In summary, we hypothesise that mixed exposure to Fe and Pb leads to osteoclast activation via the JAK-STAT signalling pathway in situ and indirectly via the gut-bone axis, resulting in bone damage. In general, our study potentially suggests that bone injury induced by mixed exposure of Fe and Pb may be related to osteoclast proliferation mediated by changes in inflammatory levels in vivo.
Collapse
Affiliation(s)
| | - Haitao Ma
- Bengbu Medical University, Bengbu 233030, China
| | | | - Liyin Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Dasheng Luo
- Department of Orthopedic Surgery, Anhui No. 2 Provincial People's Hospital, Hefei 230041, China
| | - Defu Yu
- Department of Orthopedic Surgery, Anhui No. 2 Provincial People's Hospital, Hefei 230041, China
| | - Tao Chen
- Bengbu Medical University, Bengbu 233030, China; Department of Orthopedic Surgery, Anhui No. 2 Provincial People's Hospital, Hefei 230041, China.
| |
Collapse
|
16
|
Jiang Y, Ye AH, He WG, Liu L, Gao X, Liu H, Liu WT, Ye FL, He DM, Liao JY, Wang J, He BC. Reducing PDK4 level constitutes a pivotal mechanism for glucocorticoids to impede osteoblastic differentiation through the enhancement of ferroptosis in mesenchymal stem cells. Stem Cell Res Ther 2025; 16:91. [PMID: 40001240 PMCID: PMC11863902 DOI: 10.1186/s13287-025-04186-9] [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: 07/24/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND This study mainly explores the possible role and mechanism of pyruvate dehydrogenase kinase 4 (PDK4) in the onset and development of Glucocorticoid-induced osteoporosis (GIOP), and seeks potential targets for the treatment of GIOP. METHODS Mesenchymal stem cells (MSCs) were treated with osteogenic induction medium. An in vitro osteogenic damage model was established by exposing MSCs to a high concentration (10- 6 M) of dexamethasone (DEX). Osteogenic markers were measured with real-time quantitative polymerase chain reaction, western blot, alkaline phosphatase staining, and Alizarin Red S staining. Ferroptosis markers were assessed through reactive oxygen species (ROS) fluorescent probe, transmission electron microscopy, and measurement of malondialdehyde (MDA). The potential mechanism was investigated using RT-qPCR, western blot, lysosomal probes, molecular docking, and other analytical approaches. The role of PDK4 was validated by using a GIOP rat model, micro-computed tomography and Masson's trichrome staining. RESULTS High concentrations (10- 6 M) of DEX inhibited osteogenic differentiation in C3H10T1/2 cells, and PDK4 exhibited the opposite effect. PDK4 partially reversed the osteogenic inhibitory effect of DEX both in vivo and in vitro. DEX caused mitochondrial shrinkage and disappearance of cristae in C3H10T1/2 cells, as well as an increase in total iron, ROS, MDA contents, and the level of ferroptosis key factors. These changes were partially weakened by PDK4. The ferroptosis inhibitor ferrostatin-1 partially blocked the inhibitory effect of DEX, while ferroptosis inducer RSL3 inhibited osteogenic differentiation and weakened the reversal effect of PDK4. DEX reduced the protein level of PDK4, which was partially weakened by Bafilomycin A1. The molecular docking results showed that DEX can directly bind with PDK4. CONCLUSION PDK4 can enhance the osteogenic differentiation ability of MSCs and bone mass of GIOP rats. DEX may promote the degradation of PDK4 via lysosome pathway, through which to weaken the osteogenic ability of MSCs by increasing ferroptosis. PDK4 may become a potential target for improving GIOP.
Collapse
Affiliation(s)
- Yue Jiang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Ai-Hua Ye
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wen-Ge He
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- Department of Orthropetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lu Liu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiang Gao
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Department of Orthropetics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hang Liu
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Department of Orthropetics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wen-Ting Liu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Fang-Lin Ye
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Dong-Mei He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jun-Yi Liao
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Jing Wang
- Department of Blood Transfusion, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
| | - Bai-Cheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
17
|
Zou L, Chen G, Rong Y, Tang C, Lv X, Fan Y. Three signalling pathways for iron overload in osteoporosis: a narrative review. J Orthop Surg Res 2025; 20:186. [PMID: 39979989 PMCID: PMC11844007 DOI: 10.1186/s13018-025-05588-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
Osteoporosis is a metabolic bone disease characterized by a decrease in the amount of bone tissue per unit volume and changes in bone microstructure, often resulting in bone fragility and increased susceptibility to fracture. Iron plays an important role in the normal physiological activities of human body, and its abnormal metabolism is one of the risk factors of osteoporosis. Iron overload, as an abnormality of iron metabolism, has been reported to be associated with osteoporosis in recent years. However, the mechanism of iron overload involved in the process of osteoporosis is not fully understood. In this review, we summarize what we have learned about iron overload-associated bone loss from clinical studies and animal models. Starting from the three signaling pathways of Wnt/β-catenin, BMP/SMADs, PI3K/AKT/mTOR, the mechanism of iron overload affecting the process of osteoporosis was explored, we got the conclusion that iron overload accelerates the process of osteoporosis by inhibiting normal wnt signaling, suppressing the BMP-2/SMADs pathway, down-regulating the PI3K/AKT/mTOR pathway to inhibit bone formation, and destroying the bone strength and load-bearing capacity, which providing a new direction for clinical treatment.
Collapse
Affiliation(s)
- Lingling Zou
- School of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou City, Sichuan, China
| | - Guiquan Chen
- School of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou City, Sichuan, China.
| | - Yi Rong
- School of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou City, Sichuan, China
| | - Cai Tang
- School of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou City, Sichuan, China
| | - Xingmin Lv
- School of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou City, Sichuan, China
| | - Yundong Fan
- School of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou City, Sichuan, China
| |
Collapse
|
18
|
Zhang Y, Chen C, Wu S, Nie C, Hu Y, Zhong J, Hong F. Analysis of the association between mixed exposure to multiple metals and comorbidity of osteopenia or osteoporosis: baseline data from the Chinese Multi-Ethnic Cohort study (CMEC). BMC Public Health 2025; 25:680. [PMID: 39972432 PMCID: PMC11837480 DOI: 10.1186/s12889-025-21825-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
Both osteoporosis and metal exposure are well-recognized public health concerns globally, particularly in the aging population. However, studies investigating the relationship between metal exposure and bone health conditions such as osteopenia and osteoporosis have either produced inconsistent results or are scarce, especially among the ethnic minorities in China. Herein, we correlated single-metal and metal mixture exposure with osteopenia and osteoporosis using a log-binomial regression model and quantile g-computation. In total, 9,206 ethnic Chinese individuals (Dong and Miao) aged 30-79 years were investigated in this study utilizing the baseline data from the Chinese multi-ethnic cohort study. In the single-metal exposure model, urinary concentrations of arsenic (As), cadmium (Cd), chromium (Cr), iron(Fe), mercury(Hg), and manganese (Mn) were positively associated with of osteopenia, whereas those of cobalt(Co) and zinc(Zn) concentrations were negatively associated. Additionally, urinary As, Cd, Cr, and Mn concentrations were positively associated with osteoporosis, whereas that of vanadium(V) was negatively associated. Furthermore, Quantile g-computation results indicated that metal mixture exposure was positively associated with both osteopenia and osteoporosis. Altogether, these findings suggest that simultaneous exposure to multiple metals can affect bone health, providing a theoretical basis for further studies on underlying complex mechanisms.
Collapse
Affiliation(s)
- Yuxin Zhang
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 561113, China
| | - Cheng Chen
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 561113, China
| | - Shenyan Wu
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 561113, China
| | - Chan Nie
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 561113, China
| | - Yuxin Hu
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 561113, China
| | - Jianqin Zhong
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 561113, China
| | - Feng Hong
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 561113, China.
| |
Collapse
|
19
|
Smirnova YD, Hanetseder D, Derigo L, Gasser AS, Vaglio-Garro A, Sperger S, Brunauer R, Korneeva OS, Duvigneau JC, Marolt Presen D, Kozlov AV. Osteosarcoma Cells and Undifferentiated Human Mesenchymal Stromal Cells Are More Susceptible to Ferroptosis than Differentiated Human Mesenchymal Stromal Cells. Antioxidants (Basel) 2025; 14:189. [PMID: 40002376 PMCID: PMC11852062 DOI: 10.3390/antiox14020189] [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: 10/31/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Current research suggests that promoting ferroptosis, a non-apoptotic form of cell death, may be an effective therapy for osteosarcoma, while its inhibition could facilitate bone regeneration and prevent osteoporosis. Our objective was to investigate whether the susceptibility to and regulation of ferroptosis differ between undifferentiated (UBC) and differentiated (DBC) human bone marrow stromal cells, as well as human osteosarcoma cells (MG63). Ferroptosis was induced by either inhibiting glutathione peroxidase 4 (GPX4) using RSL3 or blocking all glutathione-dependent enzymes through inhibition of the glutamate/cysteine antiporter with Erastin. Lipid peroxidation was assessed using the fluorescent probe BODIPY™581/591C11, while Ferrostatin-1 was used to inhibit ferroptosis. We demonstrate that neither Erastin nor RSL3 induces ferroptosis in DBC. However, both RSL3 and Erastin induce ferroptosis in UBC, while Erastin predominantly induces ferroptosis in MG63 cells. Our data suggest that ferroptosis induction in undifferentiated hBMSCs is primarily regulated by GPX4, whereas glutathione S-Transferase P1 (GSTP1) plays a key role in controlling ferroptosis in osteosarcoma cells. In conclusion, targeting the key pathways involved in ferroptosis across different bone cell types may improve the efficacy of cancer treatments while minimizing collateral damage and supporting regenerative processes, with minimal impact on cancer therapy.
Collapse
Affiliation(s)
- Yuliya D. Smirnova
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (Y.D.S.); (D.H.); (L.D.); (A.S.G.); (A.V.-G.); (S.S.); (R.B.); (D.M.P.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia;
| | - Dominik Hanetseder
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (Y.D.S.); (D.H.); (L.D.); (A.S.G.); (A.V.-G.); (S.S.); (R.B.); (D.M.P.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Lukas Derigo
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (Y.D.S.); (D.H.); (L.D.); (A.S.G.); (A.V.-G.); (S.S.); (R.B.); (D.M.P.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Andreas Sebastian Gasser
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (Y.D.S.); (D.H.); (L.D.); (A.S.G.); (A.V.-G.); (S.S.); (R.B.); (D.M.P.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Annette Vaglio-Garro
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (Y.D.S.); (D.H.); (L.D.); (A.S.G.); (A.V.-G.); (S.S.); (R.B.); (D.M.P.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Simon Sperger
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (Y.D.S.); (D.H.); (L.D.); (A.S.G.); (A.V.-G.); (S.S.); (R.B.); (D.M.P.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Regina Brunauer
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (Y.D.S.); (D.H.); (L.D.); (A.S.G.); (A.V.-G.); (S.S.); (R.B.); (D.M.P.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Olga S. Korneeva
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia;
| | | | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (Y.D.S.); (D.H.); (L.D.); (A.S.G.); (A.V.-G.); (S.S.); (R.B.); (D.M.P.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Andrey V. Kozlov
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (Y.D.S.); (D.H.); (L.D.); (A.S.G.); (A.V.-G.); (S.S.); (R.B.); (D.M.P.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
20
|
Li L, Liang W, Deng B, Jiang Y, Huang X, Zhang Y, Lu T, Wang L, Xu Y, Chen G. Association of dietary niacin intake with osteoporosis in the postmenopausal women in the US: NHANES 2007-2018. Front Med (Lausanne) 2025; 12:1504892. [PMID: 39975675 PMCID: PMC11835798 DOI: 10.3389/fmed.2025.1504892] [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: 10/01/2024] [Accepted: 01/23/2025] [Indexed: 02/21/2025] Open
Abstract
Background Elderly individuals with inadequate vitamin B level are at increased risk of degenerative conditions, notably cardiovascular disorders, cognitive impairments, and osteoporosis. The relationship between niacin (vitamin B3) consumption and osteoporosis risk remains a subject of debate. This study aimed to clarify the relationship between dietary niacin intake and the incidence of osteoporosis in postmenopausal women aged ≥50 years. Methods In this study, we gathered details on participants' bone mineral density, osteoporosis status, dietary niacin intake, and several other critical variables. Multivariate logistic regression models were constructed to determine the association between dietary niacin intake and the incidence of osteoporosis. Restricted cubic splines were employed to further assess the linearity and explore the shape of the dose-response associations. Additionally, we performed stratified and interaction analyses to illustrate the stability of the observed relationships across different subgroups. Results After adjusting for all covariates, there was a significant inverse association with osteoporosis (OR = 0.87; 95% CI: 0.77-0.97; p = 0.016). A negative relationship was observed between dietary niacin intake and the risk of osteoporosis (nonlinear: p = 0.672). While stratified analyses revealed some differences in the association between dietary niacin intake and osteoporosis risk, these differences were not statistically significant. Conclusion Dietary niacin intake exhibited an inverse correlation with the incidence of osteoporosis. The risk of osteoporosis was significantly reduced by 13% with every 10 mg/day increase in daily dietary niacin consumption among postmenopausal women.
Collapse
Affiliation(s)
- Li Li
- The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wankun Liang
- The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Bing Deng
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue Jiang
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaomin Huang
- The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yanlin Zhang
- The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Tianrui Lu
- The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lu Wang
- The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yunxiang Xu
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guizhen Chen
- The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
21
|
Yan CY, Gu XY, Tan SY, Mei AY, Mao JH, Dai Y, Niu J, Li WX, Kurihara H, Li YF, He RR. Lipid peroxidation inhibition by icaritin and its glycosides as a strategy to combat iron overload-induced osteoporosis in zebrafish. Food Res Int 2025; 203:115900. [PMID: 40022407 DOI: 10.1016/j.foodres.2025.115900] [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: 11/07/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 03/03/2025]
Abstract
This study provides a comprehensive evaluation of the anti-osteoporotic effects of flavonoids derived from Epimedium, including icaritin and its six glycosides-icariside I, icariside II, icariin, epimedin A, epimedin B, and epimedin C-using a zebrafish model of iron overload-induced osteoporosis. Our results demonstrate a significant increase in lipid peroxidation in zebrafish subjected to ferric ammonium citrate (FAC)-induced osteoporosis, along with impaired expression and activity of glutathione peroxidase 4 (GPX4). Treatment with ferrostatin-1, a lipid peroxide scavenger, partially alleviated the osteoporotic effects induced by FAC, implying that lipid peroxidation may play a key role in iron overload-related osteoporosis. We observed varying degrees of anti-osteoporotic activity and enhancement of osteogenic differentiation markers, such as bmp2b, runx2b, col1a1a, and alp, among icaritin and its glycosides. Notably, icaritin exhibited the most potent inhibitory effects on osteoporosis, while epimedin A and epimedin B showed enhanced efficacy compared to other glycosides, correlating closely with their ability to suppress lipid peroxidation. Additionally, through CETSA, molecular docking, and dynamic simulation studies, we identified an interaction between icaritin and GPX4, which may help stabilizing GPX4 against FAC-induced lipid peroxidation. These findings suggest that the anti-osteoporotic effects of icaritin and its glycosides are linked to their ability to suppress lipid peroxidation, offering potential therapeutic insights for managing iron overload-induced osteoporosis.
Collapse
Affiliation(s)
- Chang-Yu Yan
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of TCM/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632 China
| | - Xiao-Yuan Gu
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming 650500 China
| | - Shuo-Yan Tan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 China; State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100 China
| | - Ao-Yu Mei
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming 650500 China
| | - Jiang-Hao Mao
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming 650500 China
| | - Yi Dai
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of TCM/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632 China
| | - Jie Niu
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of TCM/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632 China
| | - Wei-Xi Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming 650500 China
| | - Hiroshi Kurihara
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of TCM/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632 China
| | - Yi-Fang Li
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of TCM/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632 China.
| | - Rong-Rong He
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of TCM/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632 China.
| |
Collapse
|
22
|
Chen H, Zhou Y, Liu Y, Zhou W, Xu L, Shang D, Ni J, Song Z. Indoxyl sulfate exacerbates alveolar bone loss in chronic kidney disease through ferroptosis. Oral Dis 2025; 31:264-277. [PMID: 38934473 DOI: 10.1111/odi.15050] [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: 02/11/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVES The purpose of this study was to determine whether indoxyl sulfate (IS) is involved in alveolar bone deterioration and to elucidate the mechanism underlying alveolar bone loss in chronic kidney disease (CKD) patients. MATERIALS AND METHODS Mice were divided into the control group, CP group (ligature-induced periodontitis), CKD group (5/6 nephrectomy), and CKD + CP group. The concentration of IS in the gingival crevicular fluid (GCF) was determined by HPLC. The bone microarchitecture was evaluated by micro-CT. MC3T3-E1 cells were stimulated with IS, and changes in mitochondrial morphology and ferroptosis-related factors were detected. RT-PCR, western blotting, alkaline phosphatase activity assays, and alizarin red S staining were utilized to assess how IS affects osteogenic differentiation. RESULTS Compared with that in the other groups, alveolar bone destruction in the CKD + CP group was more severe. IS accumulated in the GCF of mice with CKD. IS activated the aryl hydrocarbon receptor (AhR) in vitro, inhibited MC3T3-E1 cell osteogenic differentiation, caused changes in mitochondrial morphology, and activated the SLC7A11/GPX4 signaling pathway. An AhR inhibitor attenuated the aforementioned changes induced by IS. CONCLUSIONS IS activated the AhR/SLC7A11/GPX4 signaling pathway, inhibited osteogenesis in MC3T3-E1 cells, and participated in alveolar bone resorption in CKD model mice through ferroptosis.
Collapse
Affiliation(s)
- Huiwen Chen
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- 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, Shanghai, China
| | - Yining Zhou
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- 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, Shanghai, China
| | - Yingli Liu
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhou
- 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, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lina Xu
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- 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, Shanghai, China
| | - Dihua Shang
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- 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, Shanghai, China
| | - Jing Ni
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- 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, Shanghai, China
| | - Zhongchen Song
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- 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, Shanghai, China
| |
Collapse
|
23
|
Passin V, Ledesma‐Colunga MG, Altamura S, Muckenthaler MU, Baschant U, Hofbauer LC, Rauner M. Depletion of macrophages and osteoclast precursors mitigates iron overload-mediated bone loss. IUBMB Life 2025; 77:e2928. [PMID: 39555707 PMCID: PMC11611226 DOI: 10.1002/iub.2928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Iron is an essential element for physiological cellular processes, but is toxic in excess. Iron overload diseases are commonly associated with low bone mass. Increased bone resorption by osteoclasts as well as decreased bone formation by osteoblasts have been implicated in bone loss under iron overload conditions. However, the exact contribution of individual cell types has not yet been formally tested. In this study, we aimed to investigate the role of osteoclast precursors in iron overload-induced bone loss. To that end, we used clodronate liposomes to deplete phagocytic cells (including macrophages and osteoclast precursors) in male C57BL/6J mice that were exposed to ferric derisomaltose. Bone microarchitecture and bone turnover were assessed after 4 weeks. The application of clodronate resulted in the efficient depletion of circulating myeloid-lineage cells by about 70%. Depletion of osteoclast precursors mitigated iron overload-induced trabecular bone loss at the lumbar vertebrae and distal femur. While clodronate treatment led to a profound inhibition of bone turnover in control mice, it significantly reduced osteoclast numbers in iron-treated mice without further impacting the bone formation rate or serum PINP levels. Our observations suggest that even though bone formation is markedly suppressed by iron overload, osteoclasts also play a key role in iron overload-induced bone loss and highlight them as potential therapeutic targets.
Collapse
Affiliation(s)
- Vanessa Passin
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| | - Maria G. Ledesma‐Colunga
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| | - Sandro Altamura
- Department of Pediatric Hematology, Oncology and ImmunologyUniversity of HeidelbergHeidelbergGermany
- Molecular Medicine Partnership UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Martina U. Muckenthaler
- Department of Pediatric Hematology, Oncology and ImmunologyUniversity of HeidelbergHeidelbergGermany
- Molecular Medicine Partnership UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Ulrike Baschant
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| | - Lorenz C. Hofbauer
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| |
Collapse
|
24
|
Zheng Y, Sun R, Yang H, Gu T, Han M, Yu C, Chen P, Zhang J, Jiang T, Ding Y, Liang L, Quan R, Yao S, Zhao X. Aucubin Promotes BMSCs Proliferation and Differentiation of Postmenopausal Osteoporosis Patients by Regulating Ferroptosis and BMP2 Signalling. J Cell Mol Med 2025; 29:e70288. [PMID: 39823248 PMCID: PMC11740986 DOI: 10.1111/jcmm.70288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 09/02/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a chronic systemic bone metabolism disorder. Promotion in the patterns of human bone marrow mesenchymal stem cells (hBMSCs) differentiation towards osteoblasts contributes to alleviating osteoporosis. Aucubin, a natural compound isolated from the well-known herbal medicine Eucommia, was previously shown to possess various pharmacological effects. However, its effects on hBMSCs of PMOP patients are unknown. The aim of this present research was to investigate the impact and underlying process of aucubin on cell proliferation and osteogenic differentiation in hBMSCs isolated from PMOP patients. The ability of aucubin to inhibit the ferroptosis induced by erastin in hBMSCs was detected; ROS production, ferrous ion levels, SOD, MDA, and GPX activities were tested by using commercial kits. Next, ALP staining, ARS staining, RT-qPCR, RNA-sequencing, and Western blot were applied for determining the mRNA and protein expression levels associated with the osteogenesis of hBMSCs. The study also explored the involvement of BMP2/Smads signalling in aucubin promoting the osteogenesis of hBMSCs and evaluated the effects of aucubin intervention on osteoporosis using an ovariectomised rat model. The results indicated that aucubin significantly inhibited ROS generation and oxidative stress induced by erastin and protected against ferroptosis in hBMSCs. Additionally, aucubin facilitated osteogenic differentiation of hBMSCs by activating the BMP2/SMADs pathway and attenuated the progression of osteoporosis in OVX rats, suggesting a potential therapeutic benefit for postmenopausal osteoporosis (PMOP).
Collapse
Affiliation(s)
- Yang Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Research Institute of OrthopedicsThe Affiliated Jiangnan Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Rongtai Sun
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Huan Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
| | - Tianyuan Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Meichun Han
- Third Clinical Medical SchoolZhejiang Chinese Medical UniversityHangzhouChina
| | - Congcong Yu
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Pengyu Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Jianhua Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Ting Jiang
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Yangyang Ding
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Long Liang
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Renfu Quan
- Research Institute of OrthopedicsThe Affiliated Jiangnan Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- Third Clinical Medical SchoolZhejiang Chinese Medical UniversityHangzhouChina
| | - Shasha Yao
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Xing Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
25
|
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
|
26
|
Piao X, Wu X, Yan Y, Li Y, Li N, Xue L, He F. Targeting EZH2 attenuates the ferroptosis-mediated osteoblast-osteoclast imbalance in rheumatoid arthritis. Int Immunopharmacol 2024; 143:113201. [PMID: 39353382 DOI: 10.1016/j.intimp.2024.113201] [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: 06/23/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE The enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) can regulate osteogenesis and osteoclastogenesis. This study aimed to further explore the effects of EZH2 modification on ferroptosis and the osteoblast-osteoclast balance in rheumatoid arthritis (RA) in vitro and in vivo. METHODS Bone marrow mesenchymal stromal cells were transfected with EZH2 overexpression (oeEZH2) and EZH2 shRNA (shEZH2) plasmids with or without ferrostatin-1 (Fer-1) treatment and subjected to an osteoblast differentiation assay. The cells were then cocultured with bone marrow-derived macrophages and subjected to an osteoclast differentiation assay. Collagen-induced arthritis (CIA) mice were generated and injected with shEZH2 adeno-associated virus (AAV). RESULTS OeEZH2 repressed osteoblast differentiation, as reflected by decreased ALP and Alizarin Red S staining and increased OPN, RUNX2, OPG and RANKL; however, shEZH2 had the opposite effects. Besides, oeEZH2 promoted osteoblast ferroptosis, as suggested by increased MDA, Fe2+, ROS, and PTGS2 but decreased GPX4 and SLC7A11; these effects could be attenuated by Fer-1 treatment. In contrast, shEZH2 ameliorated osteoblast ferroptosis. OeEZH2 subsequently increased osteoclast differentiation, as indicated by increased TRAP+ multinucleated cells, NFATC1, CTSK, and c-FOS; however, shEZH2 had the opposite effect, except that it did not regulate CTSK. In CIA mice, shEZH2 AAV decreased the clinical symptom score, histological score of cartilage, and systemic inflammation (TNF-α and IL-6) and repressed bone ferroptosis and restored the osteoblast-osteoclast balance to some extent, as reflected by immunohistochemical staining of related markers. CONCLUSION Targeting EZH2 attenuates the ferroptosis-mediated osteoblast-osteoclast imbalance in RA, revealing its potential as a treatment target.
Collapse
Affiliation(s)
- Xuemei Piao
- Department of Rheumatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiangxiang Wu
- Department of Rheumatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yixin Yan
- Department of Internal Medicine, The Third People's Hospital of Chongming District, Shanghai 202153, China
| | - Yongming Li
- Department of Internal Medicine, The Third People's Hospital of Chongming District, Shanghai 202153, China
| | - Na Li
- Department of Internal Medicine, The Third People's Hospital of Chongming District, Shanghai 202153, China
| | - Luan Xue
- Department of Rheumatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Feng He
- The Center for Cancer Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
27
|
Chen S, Pan Y, Guo Y, Sun X, Bai X, Liu M, Wang L, Xiao J, Chen C, Ma Y. Integrative bioinformatics and experimental analysis of curcumin's role in regulating ferroptosis to combat osteoporosis. Biochem Biophys Res Commun 2024; 739:150949. [PMID: 39541922 DOI: 10.1016/j.bbrc.2024.150949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/17/2024]
Abstract
This study utilized bioinformatics and data mining techniques to explore the molecular mechanism of curcumin in treating osteoporosis (OP) through the lens of ferroptosis, thereby identifying novel therapeutic targets. The datasets GSE35958, GSE35956, GSE7429, and GSE7158 were obtained from the Gene Expression Omnibus (GEO) database. GSE35958 and GSE35956 were employed as training sets for data integration, while GSE7429 and GSE7158 served as independent validation sets. Through retrieval from the FerrDb database, iron death-related genes (FRGs) were identified, and the differentially expressed genes (DEGs) were intersected to obtain differentially expressed FRGs (DEFRGs). Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted, followed by the construction of a Protein-Protein Interaction (PPI) network to identify Hub genes. The DGIdb database was utilized to predict candidate drugs associated with the Hub genes, and molecular docking and in vitro experiments confirmed that curcumin targets the Hub genes EGFR and PTGS2.Through in vitro testing of curcumin on BMSC cells, researchers examined cell vitality, iron death, osteogenic differentiation, mineralization, and the impact on EGFR and PTGS2 levels. The analysis yielded 2212 DEGs, 484 FRGs, with 45 FDEGs at the intersection. GO analysis revealed involvement in regulating mitochondrial proteins, amino acid transport across plasma membranes, and protein ubiquitination. KEGG pathway analysis indicated associations with iron death, FoxO signaling, mTOR signaling, cell aging, osteoclast differentiation, and GSH metabolism. Utilizing the MCC algorithm, five Hub genes were identified: MAPK3, PTGS2, TGFB1, CYBB, and EGFR, showing diagnostic potential for iron death. Curcumin displayed affinity for EGFR and PTGS2, mitigating iron-induced effects on BMSCs such as increased reactive oxygen species, Fe3+ levels, and decreased mitochondrial membrane potential. Furthermore, curcumin reversed these effects, suggesting EGFR and PTGS2 as targets for curcumin to inhibit BMSC ferroptosis and potentially delay osteoporosis development. Maintaining iron homeostasis and targeting BMSC ferroptosis could offer therapeutic avenues for iron overload-induced osteoporosis.
Collapse
Affiliation(s)
- Shuangliu Chen
- School of Chinese Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Yalan Pan
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Yang Guo
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Xiaoxian Sun
- Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Xue Bai
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Mengmin Liu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Lining Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Jiřimutu Xiao
- College of Mongolian Medicine, Inner Mongolia Medical University, China
| | - Cheng Chen
- Jiangyan Hospital Affiliated to Nanjing University of Chinese Medicine, China.
| | - Yong Ma
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China; Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, China.
| |
Collapse
|
28
|
Lin S, Yincang W, Jiazhe D, Xilin X, Zhang X. Pharmacology and mechanisms of apigenin in preventing osteoporosis. Front Pharmacol 2024; 15:1486646. [PMID: 39726788 PMCID: PMC11669520 DOI: 10.3389/fphar.2024.1486646] [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: 08/26/2024] [Accepted: 11/13/2024] [Indexed: 12/28/2024] Open
Abstract
Osteoporosis (OP) stands as the most prevalent systemic skeletal condition associated with aging. The current clinical management of OP predominantly depends on anti-resorptive and anabolic agents. Nevertheless, prolonged use of some of these medications has been observed to reduce efficacy and elevate adverse effects. Given the necessity for sustained or even lifelong treatment of OP, the identification of drugs that are not only effective but also safe and cost-efficient is of utmost significance. As disease treatment paradigms continue to evolve and recent advancements in OP research come to light, certain plant-derived compounds have emerged, presenting notable benefits in the management of OP. This review primarily explores the pharmacological properties of apigenin and elucidates its therapeutic mechanisms in the context of OP. The insights provided herein aspire to offer a foundation for the judicious use of apigenin in forthcoming research, particularly within the scope of OP.
Collapse
Affiliation(s)
- Sun Lin
- Second Affiliated Hospital of Heilongjiang, University Of Chinese Medicine, Harbin, China
| | - Wang Yincang
- Second Affiliated Hospital of Heilongjiang, University Of Chinese Medicine, Harbin, China
| | - Du Jiazhe
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xu Xilin
- The Third Affiliated Hospital of Heilongjiang, University of Chinese Medicine, Harbin, China
| | - Xiaofeng Zhang
- Second Affiliated Hospital of Heilongjiang, University Of Chinese Medicine, Harbin, China
| |
Collapse
|
29
|
Jia X, Zhang G, Yu D. Application of extracellular vesicles in diabetic osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1466775. [PMID: 39720256 PMCID: PMC11666354 DOI: 10.3389/fendo.2024.1466775] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/15/2024] [Indexed: 12/26/2024] Open
Abstract
As the population ages, the occurrence of osteoporosis is becoming more common. Diabetes mellitus is one of the factors in the development of osteoporosis. Compared with the general population, the incidence of osteoporosis is significantly higher in diabetic patients. Diabetic osteoporosis (DOP) is a metabolic bone disease characterized by abnormal bone tissue structure due to hyperglycemia and insulin resistance, reduced bone strength and increased risk of fractures. This is a complex mechanism that occurs at the cellular level due to factors such as blood vessels, inflammation, and hyperglycemia and insulin resistance. Although the application of some drugs in clinical practice can reduce the occurrence of DOP, the incidence of fractures caused by DOP is still very high. Extracellular vesicles (EVs) are a new communication mode between cells, which can transfer miRNAs and proteins from mother cells to target cells through membrane fusion, thereby regulating the function of target cells. In recent years, the role of EVs in the pathogenesis of DOP has been widely demonstrated. In this article, we first describe the changes in the bone microenvironment of osteoporosis. Second, we describe the pathogenesis of DOP. Finally, we summarize the research progress and challenges of EVs in DOP.
Collapse
Affiliation(s)
- Xiaopeng Jia
- Trauma Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Gongzi Zhang
- Department of Rehabilitation Medicine, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Deshui Yu
- Trauma Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
30
|
Zhu M, Yu J. Salidroside alleviates ferroptosis in FAC-induced Age-related macular degeneration models by activating Nrf2/SLC7A11/GPX4 axis. Int Immunopharmacol 2024; 142:113041. [PMID: 39260309 DOI: 10.1016/j.intimp.2024.113041] [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: 06/29/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION Age-related macular degeneration (AMD) is a significant contributor to irreversible impairment in visual capability, particularly in its non-neovascular (dry) form. Ferroptosis, an emerging form of programmed necrosis, involves generating lipid peroxidation (LOS) through free iron and reactive oxygen species (ROS). Salidroside, a glycoside from Rhodiola rosea, known for anti-inflammatory and antioxidant properties. The research aim was exploring whether ferroptosis exists in dry AMD pathogenesis and elucidate salidroside's protective mechanisms against ferroptosis in AMD murine models and ARPE-19 cells. METHODS ARPE-19 cells were treated with varying concentrations of ferrous ammonium citrate (FAC) and salidroside. In an in vivo model, C57BL/6 mice were administered intraperitoneal injections of salidroside for 7 consecutive days, followed by an intravitreal injection (IVT) of FAC. After 7 days, the eyeballs were harvested for subsequent analyses. Ferroptosis markers were assessed using western blotting, immunofluorescence staining, and flow cytometry. To further elucidate the modulatory role of Nrf2 in ferroptosis, ARPE-19 cells were transfected with si-Nrf2. RESULTS In vitro, FAC-treated ARPE-19 cells exhibited reduced viability, decreased mitochondrial membrane potential (MMP), and accumulation of iron and lipid peroxidation (LOS) products. In vivo, FAC administration by IVT led to outer nuclear layer thinning and compromised tight junctions in RPE cells. The GPX4, Nrf2, and SLC7A11 expressions were downregulated both in vitro and in vivo. Salidroside upregulated Nrf2 and ameliorated these outcomes, but its effects were attenuated in ARPE-19 cells transfected with si-Nrf2. CONCLUSION Our study establishes that FAC induces RPE cell ferroptosis within dry AMD, and salidroside exerts therapeutic effects by triggering Nrf2/SLC7A11/GPX4 signaling axis.
Collapse
Affiliation(s)
- Meijiang Zhu
- Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China.
| | - Jing Yu
- Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China.
| |
Collapse
|
31
|
Yang Y, Zhang X, Yang Y, Gao P, Fan W, Zheng T, Yang W, Tang Y, Cai K. A two-pronged approach to inhibit ferroptosis of MSCs caused by the iron overload in postmenopausal osteoporosis and promote osseointegration of titanium implant. Bioact Mater 2024; 41:336-354. [PMID: 39161794 PMCID: PMC11331706 DOI: 10.1016/j.bioactmat.2024.07.024] [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: 03/31/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 08/21/2024] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a prevalent condition among elderly women. After menopause, women exhibit decreased iron excretion, which is prone to osteoporosis. To design a specific titanium implant for PMOP, we first analyze miRNAs and DNA characteristics of postmenopausal patients with and without osteoporosis. The results indicate that iron overload disrupts iron homeostasis in the pathogenesis of PMOP. Further experiments confirm that iron overload can cause lipid peroxidation and ferroptosis of MSCs, thus breaking bone homeostasis. Based on the findings above, we have designed a novel Ti implant coated with nanospheres of caffeic acid (CA) and deferoxamine (DFO). CA can bind on the Ti surface through the two adjacent phenolic hydroxyls and polymerize into polycaffeic acid (PCA) dimer, as well as the PCA nanospheres with the repetitive 1,4-benzodioxan units. DFO was grafted with PCA through borate ester bonds. The experimental results showed that modified Ti can inhibit the ferroptosis of MSCs in the pathological environment of PMOP and promote osseointegration in two main ways. Firstly, DFO was released under high oxidative stress, chelating with excess iron and decreasing the labile iron pool in MSCs. Meanwhile, CA and DFO activated the KEAP1/NRF2/HMOX1 pathway in MSCs and reduced the level of intracellular lipid peroxidation. So, the ferroptosis of MSCs is inhibited by promoting the SLC7A11/GSH/GPX4 pathway. Furthermore, the remained CA coating on the Ti surface could reduce the extracellular oxidative stress and glutathione level. This study offers a novel inspiration for the specific design of Ti implants in the treatment of PMOP.
Collapse
Affiliation(s)
- Yulu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xianhui Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yao Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Pengfei Gao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Wuzhe Fan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Tao Zheng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Weihu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yu Tang
- Orthopedics Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
32
|
Guo Z, Chi R, Peng Y, Sun K, Liu H, Guo F, Guo J. The Role and Interactive Mechanism of Endoplasmic Reticulum Stress and Ferroptosis in Musculoskeletal Disorders. Biomolecules 2024; 14:1369. [PMID: 39595546 PMCID: PMC11591632 DOI: 10.3390/biom14111369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/27/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Endoplasmic reticulum (ER) stress is a cellular phenomenon that arises in response to the accumulation of misfolded proteins within the ER. This process triggers the activation of a signalling pathway known as the unfolded protein response (UPR), which aims to restore ER homeostasis by reducing protein synthesis, increasing protein degradation, and promoting proper protein folding. However, excessive ER stress can perturb regular cellular function and contribute to the development of diverse pathological conditions. As is well known, ferroptosis is a kind of programmed cell death characterized by the accumulation of lipid peroxides and iron-dependent reactive oxygen species (ROS), resulting in oxidative harm to cellular structures. In recent years, there has been increasing evidence indicating that ferroptosis occurs in musculoskeletal disorders (MSDs), with emerging recognition of the complex relationship between ER stress and ferroptosis. This review presents a summary of ER stress and the ferroptosis pathway. Most importantly, it delves into the significance of ER stress in the ferroptosis process within diverse skeletal or muscle cell types. Furthermore, we highlight the potential benefits of targeting the correlation between ER stress and ferroptosis in treating degenerative MSDs.
Collapse
Affiliation(s)
- Zhou Guo
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.G.); (K.S.); (H.L.)
| | - Ruimin Chi
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Yawen Peng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- State Key Laboratory of Reproductive Medicine, The Center for Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kai Sun
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.G.); (K.S.); (H.L.)
| | - Haigang Liu
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.G.); (K.S.); (H.L.)
| | - Fengjing Guo
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.G.); (K.S.); (H.L.)
| | - Jiachao Guo
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
33
|
Bao J, Wei Y, Chen L. [Research progress on the regulatory cell death of osteoblasts in periodontitis]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:533-540. [PMID: 38803282 PMCID: PMC11528140 DOI: 10.3724/zdxbyxb-2024-0038] [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: 01/18/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024]
Abstract
Periodontitis is a chronic inflammatory disease characterized by progressive destruction of alveolar bone. The most critical mechanism underlying alveolar bone destruction is the imbalance of bone homeostasis, where osteoblast-mediated bone matrix synthesis plays an important role in regulating bone homeostasis. Regulated cell death is instrumental in both the inflammatory microenvironment and the regulation of bone homeostasis. Chronic inflammation, oxidative stress, and other factors can be directly involved in mitochondrial and death receptor-mediated signaling pathways, modulating B-cell lymphoma 2 family proteins and cysteine aspartic acid specific protease (caspase) activity, thereby affecting osteoblast apoptosis and alveolar bone homeostasis. Chronic inflammation and cellular damage induce osteoblast necroptosis via the RIPK1/RIPK3/MLKL signaling pathway, exacerbating the inflammatory response and accelerating alveolar bone destruction. Stimuli such as pathogenic microorganisms and cellular injury may also activate caspase-1-dependent or independent signaling pathways and gasdermin D family proteins, promoting osteoblast pyroptosis and releasing pro-inflammatory cytokines to mediate alveolar bone damage. Iron overload and lipid peroxidation in periodontitis can trigger ferroptosis in osteoblasts, impacting their survival and function, ultimately leading to bone homeostasis imbalance. This article focuses on the mechanism of periodontal disease affecting bone homeostasis through regulatory cell death, aiming to provide research evidence for the treatment of periodontitis and alveolar bone homeostasis imbalance.
Collapse
Affiliation(s)
- Jiaqi Bao
- Department of Periodontics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Yingming Wei
- Department of Periodontics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Lili Chen
- Department of Periodontics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
34
|
Zhou X, Jiang J, Dang J, Wang Y, Hu R, Shen C, Zhao T, Sun D, Wang G, Zhang M. Intelligent Supramolecular Modification for Implants: Endogenous Regulation of Bone Defect Repair in Osteoporosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406227. [PMID: 39166701 DOI: 10.1002/adma.202406227] [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: 05/01/2024] [Revised: 07/31/2024] [Indexed: 08/23/2024]
Abstract
Addressing osteoporosis-related bone defects, a supramolecular strategy is innovated for modifying carbon fiber reinforced polyether ether ketone (CF/PEEK) composites. By covalently attaching intelligent macromolecules via in situ RAFT polymerization, leveraging the unique pathological microenvironment in patients with iron-overloaded osteoporosis, intelligent supramolecular modified implant surface possesses multiple endogenous modulation capabilities. After implantation, surface brush-like macromolecules initially resist macrophage adhesion, thereby reducing the level of immune inflammation. Over time, the molecular chains undergo conformational changes due to Fe (III) mediated supramolecular self-assembly, transforming into mechanistic signals. These signals are then specifically transmitted to pre-osteoblast cell through the binding capacity of the KRSR short peptide at the molecular terminus, induced their osteogenic differentiation via the YAP/β-catenin signaling axis. Furthermore, osteoblasts secrete alkaline phosphatase (ALP), which significantly hydrolyzes phosphate ester bonds in surface macromolecular side groups, resulting in the release of alendronate (ALN). This process further improves the local osteoporotic microenvironment. This intelligent surface modification tailors bone repair to individual conditions, automatically realize multiple endogenous regulation once implanted, and truly realize spontaneous activation of a series of responses conducive to bone repair in vivo. It is evidenced by improved bone regeneration in iron-overloaded osteoporotic rabbits and supported by in vitro validations.
Collapse
Affiliation(s)
- Xingyu Zhou
- Engineering Research Center of Special Engineering Plastics, Ministry of Education, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Junhui Jiang
- Engineering Research Center of Special Engineering Plastics, Ministry of Education, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Junbo Dang
- Engineering Research Center of Special Engineering Plastics, Ministry of Education, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Yilong Wang
- Engineering Research Center of Special Engineering Plastics, Ministry of Education, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Ruibo Hu
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Chen Shen
- Engineering Research Center of Special Engineering Plastics, Ministry of Education, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Tianhao Zhao
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Dahui Sun
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Guibin Wang
- Engineering Research Center of Special Engineering Plastics, Ministry of Education, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Mei Zhang
- Engineering Research Center of Special Engineering Plastics, Ministry of Education, College of Chemistry, Jilin University, Changchun, 130012, China
| |
Collapse
|
35
|
Zhang Y, Hu K, Shang Z, Yang X, Cao L. Ferroptosis: Regulatory mechanisms and potential targets for bone metabolism: A review. Medicine (Baltimore) 2024; 103:e39158. [PMID: 39331895 PMCID: PMC11441915 DOI: 10.1097/md.0000000000039158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/10/2024] [Indexed: 09/29/2024] Open
Abstract
Bone homeostasis is a homeostasis process constructed by osteoblast bone formation and osteoclast bone resorption. Bone homeostasis imbalance and dysfunction are the basis for the development of various orthopedic diseases such as osteoporosis, osteoarthritis, and steroid-induced avascular necrosis of femoral head. Previous studies have demonstrated that ferroptosis can induce lipid peroxidation through the generation of reactive oxygen species, activate a number of signaling pathways, and participate in the regulation of osteoblast bone formation and osteoclast bone resorption, resulting in bone homeostasis imbalance, which is an important factor in the pathogenesis of many orthopedic diseases, but the mechanism of ferroptosis is still unknown. In recent years, it has been found that, in addition to iron metabolism and intracellular antioxidant system imbalance, organelle dysfunction is also a key factor affecting ferroptosis. This paper takes this as the starting point, reviews the latest literature reports at home and abroad, elaborates the pathogenesis and regulatory pathways of ferroptosis and the relationship between ferroptosis and various organelles, and summarizes the mechanism by which ferroptosis mediates bone homeostasis imbalance, with the aim of providing new directions for the research related to ferroptosis and new ideas for the prevention and treatment of bone and joint diseases.
Collapse
Affiliation(s)
- Yongjie Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Kangyi Hu
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhengya Shang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaorui Yang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linzhong Cao
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
- The Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
36
|
Huang L, Wang J, Yu J, Bian M, Xiang X, Han G, Chen W, Wang N, Ge J, Lu S, Zhang J. Picein alleviates oxidative stress and promotes bone regeneration in osteoporotic bone defect by inhibiting ferroptosis via Nrf2/HO-1/GPX4 pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:4066-4085. [PMID: 38727095 DOI: 10.1002/tox.24239] [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: 12/10/2023] [Revised: 02/23/2024] [Accepted: 03/14/2024] [Indexed: 06/12/2024]
Abstract
Osteoporosis (OP) can result in slower bone regeneration than the normal condition due to abnormal oxidative stress and high levels of reactive oxygen species (ROS), a condition detrimental for bone formation, making the OP-related bone healing a significant clinical challenge. As the osteogenic differentiation ability of bone marrow mesenchymal stem cells (BMSCs) is closely related to bone regeneration; currently, this study assessed the effects of Picein on BMSCs in vitro and bone regeneration in osteoporotic bone defect in vivo. Cell viability was determined by CCK-8 assay. The production of (ROS), malonaldehyde, superoxide dismutase activities, and glutathione was evaluated by using commercially available kits, and a flow cytometry analysis was adopted to detect macrophage polarization. Osteogenic capacity of BMSCs was evaluated by alkaline phosphatase (ALP) activity, ALP staining, and Alizarin red S staining. The expression of osteogenic-related proteins (OPN, Runx-2, OCN) and osteogenic-related genes (ALP, BMP-4, COL-1, and Osterix) were evaluated by Western blotting and real-time PCR (RT-PCR). In addition, proliferation, migration ability, and angiogenic capacity of human umbilical vein endothelial cells (HUVECs) were evaluated by EdU staining, scratch test, transwell assay, and tube formation assay, respectively. Angiogenic-related genes (VEGF, vWF, CD31) were also evaluated by RT-PCR. Results showed that Picein alleviated erastin-induced oxidative stress, enhanced osteogenic differentiation capacity of BMSCs, angiogenesis of HUVECs, and protects cells against ferroptosis through Nrf2/HO-1/GPX4 axis. Moreover, Picein regulate immune microenvironment by promoting the polarization of M2 macrophages in vitro. In addition, Picein also reduce the inflammation levels and promotes bone regeneration in osteoporotic bone defect in OP rat models in vivo. Altogether, these results suggested that Picein can promote bone regeneration and alleviate oxidative stress via Nrf2/HO-1/GPX4 pathway, offering Picein as a novel antioxidant agent for treating osteoporotic bone defect.
Collapse
Affiliation(s)
- Lei Huang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiayi Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jieqin Yu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xingdong Xiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guanjie Han
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weisin Chen
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ning Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Ge
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jian Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Lyamzaev KG, Huan H, Panteleeva AA, Simonyan RA, Avetisyan AV, Chernyak BV. Exogenous Iron Induces Mitochondrial Lipid Peroxidation, Lipofuscin Accumulation, and Ferroptosis in H9c2 Cardiomyocytes. Biomolecules 2024; 14:730. [PMID: 38927133 PMCID: PMC11201805 DOI: 10.3390/biom14060730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Lipid peroxidation plays an important role in various pathologies and aging, at least partially mediated by ferroptosis. The role of mitochondrial lipid peroxidation during ferroptosis remains poorly understood. We show that supplementation of exogenous iron in the form of ferric ammonium citrate at submillimolar doses induces production of reactive oxygen species (ROS) and lipid peroxidation in mitochondria that precede ferroptosis in H9c2 cardiomyocytes. The mitochondria-targeted antioxidant SkQ1 and the redox mediator methylene blue, which inhibits the production of ROS in complex I of the mitochondrial electron transport chain, prevent both mitochondrial lipid peroxidation and ferroptosis. SkQ1 and methylene blue also prevented accumulation of lipofuscin observed after 24 h incubation of cardiomyocytes with ferric ammonium citrate. Using isolated cardiac mitochondria as an in vitro ferroptosis model, it was shown that rotenone (complex I inhibitor) in the presence of ferrous iron stimulates lipid peroxidation and lipofuscin accumulation. Our data indicate that ROS generated in complex I stimulate mitochondrial lipid peroxidation, lipofuscin accumulation, and ferroptosis induced by exogenous iron.
Collapse
Affiliation(s)
- Konstantin G. Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
- The Russian Clinical Research Center for Gerontology, Ministry of Healthcare of the Russian Federation, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - He Huan
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| | - Alisa A. Panteleeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| | - Ruben A. Simonyan
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| | - Armine V. Avetisyan
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| | - Boris V. Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| |
Collapse
|
38
|
Tian B, Li X, Li W, Shi Z, He X, Wang S, Zhu X, Shi N, Li Y, Wan P, Zhu C. CRYAB suppresses ferroptosis and promotes osteogenic differentiation of human bone marrow stem cells via binding and stabilizing FTH1. Aging (Albany NY) 2024; 16:8965-8979. [PMID: 38787373 PMCID: PMC11164484 DOI: 10.18632/aging.205851] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/25/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Bone formation and homeostasis are greatly dependent on the osteogenic differentiation of human bone marrow stem cells (BMSCs). Therefore, revealing the mechanisms underlying osteogenic differentiation of BMSCs will provide new candidate therapeutic targets for osteoporosis. METHODS The osteogenic differentiation of BMSCs was measured by analyzing ALP activity and expression levels of osteogenic markers. Cellular Fe and ROS levels and cell viability were applied to evaluate the ferroptosis of BMSCs. qRT-PCR, Western blotting, and co-immunoprecipitation assays were harnessed to study the molecular mechanism. RESULTS The mRNA level of CRYAB was decreased in the plasma of osteoporosis patients. Overexpression of CRYAB increased the expression of osteogenic markers including OCN, OPN, RUNX2, and COLI, and also augmented the ALP activity in BMSCs, on the contrary, knockdown of CRYAB had opposite effects. IP-MS technology identified CRYAB-interacted proteins and further found that CRYAB interacted with ferritin heavy chain 1 (FTH1) and maintained the stability of FTH1 via the proteasome mechanism. Mechanically, we unraveled that CRYAB regulated FTH1 protein stability in a lactylation-dependent manner. Knockdown of FTH1 suppressed the osteogenic differentiation of BMSCs, and increased the cellular Fe and ROS levels, and eventually promoted ferroptosis. Rescue experiments revealed that CRYAB suppressed ferroptosis and promoted osteogenic differentiation of BMSCs via regulating FTH1. The mRNA level of FTH1 was decreased in the plasma of osteoporosis patients. CONCLUSIONS Downregulation of CRYAB boosted FTH1 degradation and increased cellular Fe and ROS levels, and finally improved the ferroptosis and lessened the osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Bo Tian
- Scientific Research Section, The First People’s Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Xiaolu Li
- Geriatric Department, The First People’s Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Weiyuan Li
- Geriatric Department, The First People’s Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Zhizhou Shi
- Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Xu He
- Geriatric Department, The First People’s Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Shengyu Wang
- Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Xun Zhu
- Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Na Shi
- Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Yan Li
- Geriatric Department, The First People’s Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Ping Wan
- Geriatric Department, The First People’s Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Chongtao Zhu
- Laser Medical Center, The First People’s Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| |
Collapse
|
39
|
Fu YF, Guo YX, Xia SH, Zhou TT, Zhao YC, Jia ZH, Zhang Y. Eldecalcitol protected osteocytes against ferroptosis of D-gal-induced senescent MLO-Y4 cells and ovariectomized mice. Exp Gerontol 2024; 189:112408. [PMID: 38521178 DOI: 10.1016/j.exger.2024.112408] [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: 01/07/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Active vitamin D analog eldecalcitol is clinically applied in treatment of postmenopausal osteoporosis. This study aims to determine the role of eldecalcitol in the protection of osteocytes from senescence and the associated ferroptosis. METHODS The MLO-Y4 osteocytes were exposed to D-gal inducing senescence. The ovariectomized (OVX) mice treated with D-gal using as an aging inducer were intraperitoneally injected with eldecalcitol. The multiplexed confocal imaging, fluorescence in situ hybridization and transmission electron microscopy were applied in assessing osteocytic properties. Immunochemical staining and immunoblotting were carried out to detect abundance and expression of molecules. RESULTS The ablation of vitamin D receptor led to a reduction in amounts of osteocytes, a loss of dendrites, an increase in mRNA expression of SASP factors and in protein expression of senescent factors, as well as changes in mRNA expression of ferroptosis-related genes (PTGS2 & RGS4). Eldecalcitol reversed senescent phenotypes of MLO-Y4 cells shown by improving cell morphology and density, decreasing β-gal-positive cell accumulation, and down-regulating protein expression (P16, P21 & P53). Eldecalcitol reduced intracellular ROS and MDA productions, elevated JC-1 aggregates, and up-regulated expression of Nrf2 and GPX4. Eldecalcitol exhibited osteopreserve effects in D-gal-induced aging OVX mice. The confocal imaging displayed its improvement on osteocytic network organization. Eldecalcitol decreased the numbers of senescent osteocytes at tibial diaphysis by SADS assay and attenuated mRNA expression of SASP factors as well as down-regulated protein expression of senescence-related factors and restored levels of ferroptotic biomarkers in osteocytes-enriched bone fraction. It reduced 4-HNE staining area, stimulated Nrf2-positive staining, and promoted nuclear translocation of Nrf2 in osteocytes of mice as well as inhibited and promoted protein expression of 4-HNE and Nrf2, respectively, in osteocytes-enriched bone fraction. CONCLUSIONS The present study revealed the ameliorative effects of eldecalcitol on senescence and the associated ferroptosis of osteocytes, contributing to its preservation against osteoporosis of D-gal-induced senescent ovariectomized mice.
Collapse
Affiliation(s)
- Yong-Fang Fu
- Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, China
| | - Yi-Xun Guo
- Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, China
| | - Shi-Hui Xia
- Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, China
| | - Ting-Ting Zhou
- Experimental Research Center, Cangzhou Hospital of Integrated TCM-WM, Cangzhou 061001, China
| | - Yun-Chao Zhao
- Experimental Research Center, Cangzhou Hospital of Integrated TCM-WM, Cangzhou 061001, China
| | - Zhen-Hua Jia
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, China.
| | - Yan Zhang
- Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, China.
| |
Collapse
|
40
|
Huang L, Zhang S, Bian M, Xiang X, Xiao L, Wang J, Lu S, Chen W, Zhang C, Mo G, Jiang L, Li Y, Zhang J. Injectable, anti-collapse, adhesive, plastic and bioactive bone graft substitute promotes bone regeneration by moderating oxidative stress in osteoporotic bone defect. Acta Biomater 2024; 180:82-103. [PMID: 38621599 DOI: 10.1016/j.actbio.2024.04.016] [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: 11/29/2023] [Revised: 03/05/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
The treatment of osteoporotic bone defect remains a big clinical challenge because osteoporosis (OP) is associated with oxidative stress and high levels of reactive oxygen species (ROS), a condition detrimental for bone formation. Anti-oxidative nanomaterials such as selenium nanoparticles (SeNPs) have positive effect on osteogenesis owing to their pleiotropic pharmacological activity which can exert anti-oxidative stress functions to prevent bone loss and facilitate bone regeneration in OP. In the current study a strategy of one-pot method by introducing Poly (lactic acid-carbonate) (PDT) and β-Tricalcium Phosphate (β-TCP) with SeNPs, is developed to prepare an injectable, anti-collapse, shape-adaptive and adhesive bone graft substitute material (PDT-TCP-SE). The PDT-TCP-SE bone graft substitute exhibits sufficient adhesion in biological microenvironments and osteoinductive activity, angiogenic effect and anti-inflammatory as well as anti-oxidative effect in vitro and in vivo. Moreover, the PDT-TCP-SE can protect BMSCs from erastin-induced ferroptosis through the Sirt1/Nrf2/GPX4 antioxidant pathway, which, in together, demonstrated the bone graft substitute material as an emerging biomaterial with potential clinical application for the future treatment of osteoporotic bone defect. STATEMENT OF SIGNIFICANCE: Injectable, anti-collapse, adhesive, plastic and bioactive bone graft substitute was successfully synthesized. Incorporation of SeNPs with PDT into β-TCP regenerated new bone in-situ by moderating oxidative stress in osteoporotic bone defects area. The PDT-TCP-SE bone graft substitute reduced high ROS levels in osteoporotic bone defect microenvironment. The bone graft substitute could also moderate oxidative stress and inhibit ferroptosis via Sirt1/Nrf2/GPX4 pathway in vitro. Moreover, the PDT-TCP-SE bone graft substitute could alleviate the inflammatory environment and promote bone regeneration in osteoporotic bone defect in vivo. This biomaterial has the advantages of simple synthesis, biocompatibility, anti-collapse, injectable, and regulation of oxidative stress level, which has potential application value in bone tissue engineering.
Collapse
Affiliation(s)
- Lei Huang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shihao Zhang
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xingdong Xiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lan Xiao
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia
| | - Jiayi Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weisin Chen
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cheng Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guokang Mo
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Libo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yulin Li
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| | - Jian Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
41
|
Chen Y, Zhao W, Hu A, Lin S, Chen P, Yang B, Fan Z, Qi J, Zhang W, Gao H, Yu X, Chen H, Chen L, Wang H. Type 2 diabetic mellitus related osteoporosis: focusing on ferroptosis. J Transl Med 2024; 22:409. [PMID: 38693581 PMCID: PMC11064363 DOI: 10.1186/s12967-024-05191-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/12/2024] [Indexed: 05/03/2024] Open
Abstract
With the aging global population, type 2 diabetes mellitus (T2DM) and osteoporosis(OP) are becoming increasingly prevalent. Diabetic osteoporosis (DOP) is a metabolic bone disorder characterized by abnormal bone tissue structure and reduced bone strength in patients with diabetes. Studies have revealed a close association among diabetes, increased fracture risk, and disturbances in iron metabolism. This review explores the concept of ferroptosis, a non-apoptotic cell death process dependent on intracellular iron, focusing on its role in DOP. Iron-dependent lipid peroxidation, particularly impacting pancreatic β-cells, osteoblasts (OBs) and osteoclasts (OCs), contributes to DOP. The intricate interplay between iron dysregulation, which comprises deficiency and overload, and DOP has been discussed, emphasizing how excessive iron accumulation triggers ferroptosis in DOP. This concise overview highlights the need to understand the complex relationship between T2DM and OP, particularly ferroptosis. This review aimed to elucidate the pathogenesis of ferroptosis in DOP and provide a prospective for future research targeting interventions in the field of ferroptosis.
Collapse
Affiliation(s)
- Yili Chen
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wen Zhao
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510006, China
| | - An Hu
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510006, China
| | - Shi Lin
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510006, China
| | - Ping Chen
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Bing Yang
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhirong Fan
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ji Qi
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wenhui Zhang
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huanhuan Gao
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiubing Yu
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Haiyun Chen
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Luyuan Chen
- Stomatology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 510086, China.
| | - Haizhou Wang
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
42
|
Lei Z, Liang H, Sun W, Chen Y, Huang Z, Yu B. A biodegradable PVA coating constructed on the surface of the implant for preventing bacterial colonization and biofilm formation. J Orthop Surg Res 2024; 19:175. [PMID: 38459593 PMCID: PMC10921624 DOI: 10.1186/s13018-024-04662-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/02/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Bone implant infections pose a critical challenge in orthopedic surgery, often leading to implant failure. The potential of implant coatings to deter infections by hindering biofilm formation is promising. However, a shortage of cost-effective, efficient, and clinically suitable coatings persists. Polyvinyl alcohol (PVA), a prevalent biomaterial, possesses inherent hydrophilicity, offering potential antibacterial properties. METHODS This study investigates the PVA solution's capacity to shield implants from bacterial adhesion, suppress bacterial proliferation, and thwart biofilm development. PVA solutions at concentrations of 5%, 10%, 15%, and 20% were prepared. In vitro assessments evaluated PVA's ability to impede bacterial growth and biofilm formation. The interaction between PVA and mCherry-labeled Escherichia coli (E. coli) was scrutinized, along with PVA's therapeutic effects in a rat osteomyelitis model. RESULTS The PVA solution effectively restrained bacterial proliferation and biofilm formation on titanium implants. PVA solution had no substantial impact on the activity or osteogenic potential of MC3T3-E1 cells. Post-operatively, the PVA solution markedly reduced the number of Staphylococcus aureus and E. coli colonies surrounding the implant. Imaging and histological scores exhibited significant improvements 2 weeks post-operation. Additionally, no abnormalities were detected in the internal organs of PVA-treated rats. CONCLUSIONS PVA solution emerges as an economical, uncomplicated, and effective coating material for inhibiting bacterial replication and biofilm formation on implant surfaces, even in high-contamination surgical environments.
Collapse
Affiliation(s)
- Zhonghua Lei
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Department of Orthopedics, The Sixth Peoples Hospital of Huizhou, Huizhou, 516211, China
| | - Haifeng Liang
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Department of Orthopedics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Wei Sun
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yan Chen
- Ultrasound Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Zhi Huang
- Institute of Biomedical Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410083, China.
| | - Bo Yu
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
43
|
Jiang Z, Qi G, He X, Yu Y, Cao Y, Zhang C, Zou W, Yuan H. Ferroptosis in Osteocytes as a Target for Protection Against Postmenopausal Osteoporosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307388. [PMID: 38233202 PMCID: PMC10966575 DOI: 10.1002/advs.202307388] [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: 10/04/2023] [Revised: 12/18/2023] [Indexed: 01/19/2024]
Abstract
Ferroptosis is a necrotic form of iron-dependent regulatory cell death. Estrogen withdrawal can interfere with iron metabolism, which is responsible for the pathogenesis of postmenopausal osteoporosis (PMOP). Here, it is demonstrated that estrogen withdrawal induces iron accumulation in the skeleton and the ferroptosis of osteocytes, leading to reduced bone mineral density. Furthermore, the facilitatory effect of ferroptosis of osteocytes is verified in the occurrence and development of postmenopausal osteoporosis is associated with over activated osteoclastogenesis using a direct osteocyte/osteoclast coculture system and glutathione peroxidase 4 (GPX4) knockout ovariectomized mice. In addition, the nuclear factor erythroid derived 2-related factor-2 (Nrf2) signaling pathway is confirmed to be a crucial factor in the ferroptosis of osteocytic cells. Nrf2 regulates the expression of nuclear factor kappa-B ligand (RANKL) by regulating the DNA methylation level of the RANKL promoter mediated by DNA methyltransferase 3a (Dnmt3a), which is as an important mechanism in osteocytic ferroptosis-mediated osteoclastogenesis. Taken together, this data suggests that osteocytic ferroptosis is involved in PMOP and can be targeted to tune bone homeostasis.
Collapse
Affiliation(s)
- Zengxin Jiang
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Guobin Qi
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Xuecheng He
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yifan Yu
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yuting Cao
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Changqing Zhang
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Weiguo Zou
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
- State Key Laboratory of Cell BiologyCAS Center for Excellence in Molecular Cell SciencesShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
| | - Hengfeng Yuan
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| |
Collapse
|
44
|
Shen Y, Zhang Y, Wang Q, Jiang B, Jiang X, Luo B. MicroRNA-877-5p promotes osteoblast differentiation by targeting EIF4G2 expression. J Orthop Surg Res 2024; 19:134. [PMID: 38342889 PMCID: PMC10860299 DOI: 10.1186/s13018-023-04396-y] [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: 07/17/2023] [Accepted: 11/20/2023] [Indexed: 02/13/2024] Open
Abstract
Stimulating bone formation potentially suggests therapeutics for orthopedic diseases including osteoporosis and osteoarthritis. Osteoblasts are key to bone remodeling because they act as the only bone-forming cells. miR-877-5p has a chondrocyte-improving function in osteoarthritis, but its effect on osteoblast differentiation is unknown. Here, miR-877-5p-mediated osteoblast differentiation was studied. Real-time reverse transcriptase-polymerase chain reaction was performed to measure miR-877-5p expression during the osteogenic differentiation of MC3T3-E1 cells. Osteoblast markers, including alkaline phosphatase (ALP), collagen type I a1 chain, and osteopontin, were measured and detected by alizarin red staining and ALP staining. Potential targets of miR-877-5p were predicted from three different algorithms: starBase ( http://starbase.sysu.edu.cn/ ), PITA ( http://genie.weizmann.ac.il/pubs/mir07/mir07_data.html ), and miRanda ( http://www.microrna.org/microrna/home.do ). It was further verified by dual luciferase reporter gene assay. The experimental results found that miR-877-5p was upregulated during the osteogenic differentiation of MC3T3-E1 cells. Overexpression of miR-877-5p promoted osteogenic differentiation, which was characterized by increased cell mineralization, ALP activity, and osteogenesis-related gene expression. Knockdown of miR-877-5p produced the opposite result. Dual luciferase reporter gene assay showed that miR-877-5p directly targeted eukaryotic translation initiation factor 4γ2 (EIF4G2). Overexpression of EIF4G2 inhibited osteogenic differentiation and reversed the promoting effect of overexpression of miR-135-5p on osteogenic differentiation. These results indicate that miR-877-5p might have a therapeutic application related to its promotion of bone formation through targeting EIF4G2.
Collapse
Affiliation(s)
- YingChao Shen
- Department of Orthopaedics, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, No. 6 Huanghe Road, ChangShu City, 215500, China
| | - Yang Zhang
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu City, 215500, Jiangsu, China
| | - Qiang Wang
- Department of Orthopaedics, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, No. 6 Huanghe Road, ChangShu City, 215500, China
| | - Bo Jiang
- Department of Hand and Foot Surgery, The Second Affiliated Hospital of Soochow University, Jiangsu Province, No. 1055 Sanxiang Road, Suzhou City, 215004, China.
| | - XiaoWei Jiang
- Department of Orthopaedics, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, No. 6 Huanghe Road, ChangShu City, 215500, China.
| | - Bin Luo
- Department of Orthopaedics, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, No. 6 Huanghe Road, ChangShu City, 215500, China
| |
Collapse
|
45
|
Li P, Wang Y, Yan Q, Yang Y, Zhu R, Ma J, Chen Y, Liu H, Zhang Z. Fructus Ligustri Lucidi inhibits ferroptosis in ovariectomy‑induced osteoporosis in rats via the Nrf2/HO‑1 signaling pathway. Biomed Rep 2024; 20:27. [PMID: 38259585 PMCID: PMC10801352 DOI: 10.3892/br.2023.1715] [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: 09/18/2023] [Accepted: 11/20/2023] [Indexed: 01/24/2024] Open
Abstract
Postmenopausal osteoporosis (PMOP) has increased in prevalence in recent years, thus researchers have evaluated alternative medicine therapies. Fructus Ligustri Lucidi (FLL) can inhibit bone loss, and ferroptosis serves an important role in osteoporosis. Therefore, the present study assessed the presence of ferroptosis in PMOP and whether FLL could inhibit ferroptosis to improve bone microstructure in ovariectomized rats. Ovariectomized rats were treated with FLL (1.56 g/kg/day) for 12 weeks. Micro-CT was performed to evaluate the bone microstructure and bone mineral density. Western blotting and reverse transcription-quantitative PCR were performed to assess the relative expression levels of proteins and mRNA. Subsequently, malondialdehyde (MDA) and Fe2+ assay kits were used to quantify the MDA and Fe2+ content, respectively. The results demonstrated that ovariectomy (OVX) resulted in iron overload and the accumulation of lipid peroxide. Furthermore, the expression of key factors that inhibited ferroptosis, glutathione peroxidase 4 and solute carrier family 7 member 11 was significantly downregulated in ovariectomized rats, which was significantly reversed by FLL treatment. Furthermore, bone formation was assessed using the expression of osteogenesis-related genes, runt-related transcription factor 2 and osterix, which revealed significantly higher levels in FLL-treated rats compared with ovariectomized rats. The levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) were also significantly recovered following FLL treatment. In the present study, OVX of postmenopausal osteoporotic rats was found to induce ferroptosis by enhancing lipid peroxidation and Fe2+ levels. FLL significantly suppressed ferroptosis, protected the osteogenic ability of ovariectomized rats and promoted the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Pei Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Yuhan Wang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Qiqi Yan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Ying Yang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Ruyuan Zhu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Jiayi Ma
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Yanjing Chen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Haixia Liu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Zhiguo Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| |
Collapse
|
46
|
Jiang J, Zhao B, Xiao J, Shi L, Shang W, Shu Y, Zhao Z, Shen J, Xu J, Cai H. Exploring the boost of steaming with wine on Ligustri Lucidi Fructus in treating postmenopausal osteoporosis based on superior "multi-component structure" and iron/bone metabolism coregulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155275. [PMID: 38142661 DOI: 10.1016/j.phymed.2023.155275] [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: 05/26/2023] [Revised: 10/07/2023] [Accepted: 12/10/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Clinical studies indicated that postmenopausal osteoporosis (PMOP) often accompanied by iron overload risk factor, which exacerbated bone metabolism disorders and accelerated PMOP. Previous research found that multicomponent in Ligustri Lucidi Fructus (FLL) or wine-steamed FLL (WFLL) acted on the common targets of iron overload and PMOP simultaneously, which indicated that FLL and WFLL probably regulated iron/bone metabolism dually. Additionally, WFLL had more superior effect according to the theory of Chinese medicine for thousands of years. PURPOSE To reveal the "superior multi-component structure (SMCS)" and its molecular mechanisms in parallelly down-regulating iron overload and rescuing bone metabolism by WFLL. DESIGNS AND METHODS HPLC fingerprinting was established to compare the chemical profiles of FLL and WFLL; Then, the chemical compositions and quality markers of FLL and WFLL were analyzed by UPLC-Orbitrap-MS/MS coupled with OPLS-DA; the dynamic contents of quality markers and the multi-component structure at different wine steaming times (WST) were simultaneously determined by HPLC-DAD. Meanwhile, the dynamic efficacy of FLL at different WST were hunt by systematic zebrafish model. Subsequently, potential mechanism of WFLL in treating PMOP accompanied with iron overload was obtained from network pharmacology (NP) and molecular docking (MD). Finally, zebrafish and ovariectomy rat model were carried out to validate this potential mechanism. RESULTS HPLC fingerprints similarity of 15 batches in FLL and WFLL were among 0.9-1.0. 126 compositions were identified, including 58 iridoids, 25 terpenes, 30 phenylethanoids, 7 flavonoids and 6 others. 20 quality markers associated with WFLL was revealed, and the ratio of phenylethanols: Iridoids: Triterpenes (P/I/T) was converted from 1: 15: 4.5 to 1: 0.8: 0.9 during steaming (0 - 24 h) calculated by the quantification of 11 quality markers; the bone mineralization and motor performance of zebrafish larvae indicated that the optimum efficacy of WFLL at 12 h (p < 0.05) in which the SMCS of P/I/T was converted to 1: 4: 1.8. NP discovered that BMP-Smad pathway is one of the potential mechanisms of FLL in anti PMOP and then regulated bone formation and iron overload simultaneously. MD revealed that 17 active ingredients and 10 core targets genes could spontaneously bind with appropriate affinity. Rats model verified that FLL and WFLL significantly reversed PMOP, based on the improvement in bone formation indexes (ALP, OPG, OGN), iron metabolism indicators (hepcidin, ferritin), bone microstructure (BMD, BV/TV, Tb. Th, Tb. N); Moreover, WFLL significant enhanced reversal effect in anti-PMOP compared to FLL (p < 0.05). FLL and WFLL increased genes and proteins expression (Hep, BMP-6, p-Smad1/5, Smad4) related to BMP-Smad pathway compared with model group, and WFLL was more superior than FLL (p< 0.05). CONCLUSION The SMCS of FLL was optimized by wine-steam, WFLL represented a dual effect in downregulating iron overload and promoting bone formation, and the BMP-Smad pathway is one of the potential molecular mechanisms.
Collapse
Affiliation(s)
- Jun Jiang
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu 212013, China; Department of TCM, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
| | - Baixiu Zhao
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Jianpeng Xiao
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Liang Shi
- Nanjing first hospital, No.68 Changle Road, Qinhuai District, Nanjing, Jiangsu 210006, China
| | - Wei Shang
- Department of TCM, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
| | - Ye Shu
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Zhiming Zhao
- Department of TCM, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Junyi Shen
- Department of TCM, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jingjuan Xu
- Department of TCM, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Hui Cai
- Department of TCM, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
| |
Collapse
|
47
|
Zhang Y, Qu Z, Zhao Y, Zhang B, Gong Y, Wang X, Gao X, Wang D, Yan L. The Therapeutic Effect of Natural Compounds on Osteoporosis through Ferroptosis. Curr Med Chem 2024; 31:2629-2648. [PMID: 37817519 DOI: 10.2174/0109298673258420230919103405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/22/2023] [Accepted: 08/18/2023] [Indexed: 10/12/2023]
Abstract
Ferroptosis is a newly discovered non-apoptotic cell death whose key is lipid peroxidation. It has been reported that ferroptosis is involved in the occurrence and development of tumors and nervous system and musculoskeletal diseases. Cellular ferroptosis contributes to the imbalance of bone homeostasis and is involved in the development of osteoporosis; however, the detailed mechanism of which is still unclear though it may provide a new direction for anti-osteoporosis. The current drugs used in the treatment of osteoporosis, such as bisphosphonates and teriparatide, have many side effects, increasing people's search for natural compounds to treat osteoporosis. This review paper briefly summarizes the current research regarding the mechanisms of ferroptosis and natural anti-osteoporosis compounds targeting its pathway.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zechao Qu
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yiwei Zhao
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bo Zhang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yining Gong
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaohui Wang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiangcheng Gao
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dong Wang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Yan
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
48
|
Wang W, Zhang H, Sandai D, Zhao R, Bai J, Wang Y, Wang Y, Zhang Z, Zhang HL, Song ZJ. ATP-induced cell death: a novel hypothesis for osteoporosis. Front Cell Dev Biol 2023; 11:1324213. [PMID: 38161333 PMCID: PMC10755924 DOI: 10.3389/fcell.2023.1324213] [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: 10/19/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
ATP-induced cell death has emerged as a captivating realm of inquiry with profound ramifications in the context of osteoporosis. This study unveils a paradigm-shifting hypothesis that illuminates the prospective involvement of ATP-induced cellular demise in the etiology of osteoporosis. Initially, we explicate the morphological attributes of ATP-induced cell death and delve into the intricacies of the molecular machinery and regulatory networks governing ATP homeostasis and ATP-induced cell death. Subsequently, our focus pivots towards the multifaceted interplay between ATP-induced cellular demise and pivotal cellular protagonists, such as bone marrow-derived mesenchymal stem cells, osteoblasts, and osteoclasts, accentuating their potential contributions to secondary osteoporosis phenotypes, encompassing diabetic osteoporosis, glucocorticoid-induced osteoporosis, and postmenopausal osteoporosis. Furthermore, we probe the captivating interplay between ATP-induced cellular demise and alternative modalities of cellular demise, encompassing apoptosis, autophagy, and necroptosis. Through an all-encompassing inquiry into the intricate nexus connecting ATP-induced cellular demise and osteoporosis, our primary goal is to deepen our comprehension of the underlying mechanisms propelling this malady and establish a theoretical bedrock to underpin the development of pioneering therapeutic strategies.
Collapse
Affiliation(s)
- Wei Wang
- College of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Haolong Zhang
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Doblin Sandai
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Rui Zhao
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jinxia Bai
- College of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yanfei Wang
- College of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yong Wang
- Pathology Center, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Zhongwen Zhang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Hao-Ling Zhang
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Zhi-Jing Song
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
49
|
Yang Y, Jiang Y, Qian D, Wang Z, Xiao L. Prevention and treatment of osteoporosis with natural products: Regulatory mechanism based on cell ferroptosis. J Orthop Surg Res 2023; 18:951. [PMID: 38082321 PMCID: PMC10712195 DOI: 10.1186/s13018-023-04448-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
CONTEXT With the development of society, the number of patients with osteoporosis is increasing. The prevention and control of osteoporosis has become a serious and urgent issue. With the continuous progress of biomedical research, ferroptosis has attracted increased attention. However, the pathophysiology and mechanisms of ferroptosis and osteoporosis still need further study. Natural products are widely used in East Asian countries for osteoporosis prevention and treatment. OBJECTIVE In this paper, we will discuss the basic mechanisms of ferroptosis, the relationship between ferroptosis and osteoclasts and osteoblasts, and in vitro and in vivo studies of natural products to prevent osteoporosis by interfering with ferroptosis. METHODS This article takes ferroptosis, natural products, osteoporosis, osteoblasts and osteoclast as key words. Retrieve literature from 2012 to 2023 indexed in databases such as PubMed Central, PubMed, Web of Science, Scopus and ISI. RESULTS Ferroptosis has many regulatory mechanisms, including the system XC -/GSH/GPX4, p62/Keap1/Nrf2, FSP1/NAD (P) H/CoQ10, P53/SAT1/ALOX15 axes etc. Interestingly, we found that natural products, such as Artemisinin, Biochanin A and Quercetin, can play a role in treating osteoporosis by promoting ferroptosis of osteoclast and inhibiting ferroptosis of osteoblasts. CONCLUSIONS Natural products have great potential to regulate OBs and OCs by mediating ferroptosis to prevent and treat osteoporosis, and it is worthwhile to explore and discover more natural products that can prevent and treat osteoporosis.
Collapse
Affiliation(s)
- Yunshang Yang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China
- Department of Orthopedics, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China
| | - Yifan Jiang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China
| | - Daoyi Qian
- Department of Orthopedics, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China
| | - Zhirong Wang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China.
- Department of Orthopedics, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China.
| | - Long Xiao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China.
- Department of Orthopedics, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China.
| |
Collapse
|
50
|
Piñera-Avellaneda D, Buxadera-Palomero J, Ginebra MP, Rupérez E, Manero JM. Gallium-doped thermochemically treated titanium reduces osteoclastogenesis and improves osteodifferentiation. Front Bioeng Biotechnol 2023; 11:1303313. [PMID: 38144539 PMCID: PMC10748490 DOI: 10.3389/fbioe.2023.1303313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Excessive bone resorption is one of the main causes of bone homeostasis alterations, resulting in an imbalance in the natural remodeling cycle. This imbalance can cause diseases such as osteoporosis, or it can be exacerbated in bone cancer processes. In such cases, there is an increased risk of fractures requiring a prosthesis. In the present study, a titanium implant subjected to gallium (Ga)-doped thermochemical treatment was evaluated as a strategy to reduce bone resorption and improve osteodifferentiation. The suitability of the material to reduce bone resorption was proven by inducing macrophages (RAW 264.7) to differentiate to osteoclasts on Ga-containing surfaces. In addition, the behavior of human mesenchymal stem cells (hMSCs) was studied in terms of cell adhesion, morphology, proliferation, and differentiation. The results proved that the Ga-containing calcium titanate layer is capable of inhibiting osteoclastogenesis, hypothetically by inducing ferroptosis. Furthermore, Ga-containing surfaces promote the differentiation of hMSCs into osteoblasts. Therefore, Ga-containing calcium titanate may be a promising strategy for patients with fractures resulting from an excessive bone resorption disease.
Collapse
Affiliation(s)
- David Piñera-Avellaneda
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Judit Buxadera-Palomero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - Elisa Rupérez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - José María Manero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| |
Collapse
|