1
|
Liu H, Li M, Deng Y, Hou Y, Hou L, Zhang X, Zheng Z, Guo F, Sun K. The Roles of DMT1 in Inflammatory and Degenerative Diseases. Mol Neurobiol 2025; 62:6317-6332. [PMID: 39775481 DOI: 10.1007/s12035-025-04687-x] [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: 04/01/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Iron homeostasis is critical for multiple physiological and pathological processes. DMT1, a core iron transporter, is expressed in almost all cells and organs and altered in response to various conditions, whereas, there is few reviews focusing on DMT1 in diseases associated with aberrant iron metabolism. Based on available knowledge, this review described a full view of DMT1 and summarized the roles of DMT1 and DMT1-mediated iron metabolism in the onset and development of inflammatory and degenerative diseases. This review also provided an overview of DMT1-related treatment in these disorders, highlighting its therapeutic potential in chronic inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Haigang Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Mi Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yi Deng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yanjun Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Liangcai Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xiong Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zehang Zheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
2
|
Li M, Wei CB, Li HF, He K, Bai RJ, Zhang FJ. Osteopontin inhibits autophagy via CD44 and avβ3 integrin and promotes cell proliferation in osteoarthritic fibroblast-like synoviocytes. BMC Musculoskelet Disord 2025; 26:274. [PMID: 40102843 PMCID: PMC11916941 DOI: 10.1186/s12891-025-08509-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/07/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is closely related to aging, and autophagy is implicated in the retardation of aging. Activated synoviocytes play important roles in OA; the synoviocytes could produce osteopontin (OPN) and its main receptors CD44 and integrin, which are all involved in OA. The purpose of this study is to investigate whether OPN has an effect on autophagy in osteoarthritic synoviocytes. METHODS We cultured human OA fibroblast-like synoviocytes (FLS) and treated them with rhOPN and antibodies against CD44 and CD51/61 (αvβ3 integrin) or isotype IgG to block the interaction between receptors and ligands. Infection with lentivirus mRFP-GFP-LC3, laser confocal imaging and Western blotting were used to determine changes in the expression of autophagy markers, and cell proliferation of FLS was assessed with a CCK-8 assay. RESULTS Our results showed the expression level of autophagy marker protein LC3 II and the mRFP-GFP-LC3 puncta were significantly decreased after treatment with rhOPN when compared with the control group, when the FLS were incubated with antibodies against CD44 or CD51/61 (αvβ3 integrin) or with control isotype IgG for 1 h, followed by rhOPN treatment for 48 h, rhOPN could suppress the relative expression of LC3 II and Beclin1 via integrin and CD44 in the FLS, CCK-8 assay also showed that rhOPN significantly increased the cell proliferation and viability of FLS. CONCLUSIONS OPN could inhibit autophagy via CD44 and αvβ3 integrin and promote the proliferation of FLS, playing an important role in OA synovitis.
Collapse
Affiliation(s)
- Min Li
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Chang-Bao Wei
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Hai-Feng Li
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Ke He
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Rui-Jun Bai
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China.
| | - Fang-Jie Zhang
- Department of Emergency Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), No.87 Xiangya Road, Changsha, Hunan, 410008, China.
| |
Collapse
|
3
|
Xu J, Gu J, Pei W, Zhang Y, Wang L, Gao J. The role of lysosomal membrane proteins in autophagy and related diseases. FEBS J 2024; 291:3762-3785. [PMID: 37221945 DOI: 10.1111/febs.16820] [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: 02/14/2023] [Revised: 04/12/2023] [Accepted: 05/15/2023] [Indexed: 05/25/2023]
Abstract
As a self-degrading and highly conserved survival mechanism, autophagy plays an important role in maintaining cell survival and recycling. The discovery of autophagy-related (ATG) genes has revolutionized our understanding of autophagy. Lysosomal membrane proteins (LMPs) are important executors of lysosomal function, and increasing evidence has demonstrated their role in the induction and regulation of autophagy. In addition, the functional dysregulation of the process mediated by LMPs at all stages of autophagy is closely related to neurodegenerative diseases and cancer. Here, we review the role of LMPs in autophagy, focusing on their roles in vesicle nucleation, vesicle elongation and completion, the fusion of autophagosomes and lysosomes, and degradation, as well as their broad association with related diseases.
Collapse
Affiliation(s)
- Jiahao Xu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Jing Gu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
| | - Wenjun Pei
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Yao Zhang
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Lizhuo Wang
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Jialin Gao
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Anhui Provincial College Key Laboratory of Non-coding RNA Transformation Research on Critical Diseases, Wannan Medical College, Wuhu, China
| |
Collapse
|
4
|
Chen R, Gong K, Chen W, Chen Z, Zhang L, Tang Y, Li Y, Zhou S. Association of dietary carbohydrate intake with bone mineral density, osteoporosis and fractures among adults without diabetes: Evidence from National Health and Nutrition Examination Survey. Heliyon 2024; 10:e35566. [PMID: 39170357 PMCID: PMC11336747 DOI: 10.1016/j.heliyon.2024.e35566] [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: 01/23/2024] [Revised: 07/23/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Background The impact of dietary carbohydrate intake on bone health remains a subject of controversy, potentially influenced by individuals with diabetic osteoporosis who exhibit normal or elevated bone mineral density (BMD). The cross-sectional study was conducted to explore the association between carbohydrate intake and BMD, osteoporosis and fractures among adults without diabetes, based on the National health and nutrition examination survey (NHANES). Methods Participants were from the NHANES 2005-2010, excluding individuals with diabetes and those with incomplete data. The association between carbohydrate intake and BMD was analyzed using Spearman correlation, linear regression analysis and subgroup analysis, respectively. The association between carbohydrate intake and osteoporosis/fractures was analyzed using weighted logistic regression analysis. Results A total of 7275 adult participants were included and their dietary carbohydrate intake was inversely associated with BMD in the total femur [β = -0.20 95%CI (-0.30, -0.10); p < 0.001], femoral neck [β = -0.10 95%CI (-0.20, -0.00); p = 0.002], and lumbar spine [β = -0.10 95%CI (-0.20, -0.00); p = 0.004]. Stratified analysis indicated that individuals aged 65 and over, women, and non-Hispanic whites were more likely to have lower BMD. Furthermore, a higher intake of dietary carbohydrates was associated with an increased risk of osteoporosis [OR = 1.001 95%CI (1.001, 1.001); p < 0.001] and fractures at the hip [OR = 1.005 95%CI (1.005, 1.005); p < 0.001], wrist [OR = 1.001 95%CI (1.001, 1.001), p < 0.001], and spine [OR = 1.003 95%CI(1.003, 1.003); p < 0.001]. Conclusions A higher carbohydrate diet is associated with lower BMD and a higher risk of osteoporosis and fractures among adults without diabetes, and a higher carbohydrate consumption show a stronger effect in individuals aged 65 and over, women, and non-Hispanic whites.
Collapse
Affiliation(s)
- Ran Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 400042, PR China
| | - Kai Gong
- Department of Orthopaedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan Province, 610500, PR China
| | - Wei Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 400042, PR China
| | - Zongfeng Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 400042, PR China
| | - Lianyang Zhang
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 400042, PR China
| | - Ying Tang
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 400042, PR China
| | - Yang Li
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 400042, PR China
| | - Siru Zhou
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 400042, PR China
| |
Collapse
|
5
|
Wang T, Xiong K, He Y, Feng B, Guo L, Gu J, Zhang M, Wang H, Wu X. Chronic pancreatitis-associated metabolic bone diseases: epidemiology, mechanisms, and clinical advances. Am J Physiol Endocrinol Metab 2024; 326:E856-E868. [PMID: 38656128 DOI: 10.1152/ajpendo.00113.2024] [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: 03/19/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Chronic pancreatitis (CP) is a progressive inflammatory disease with an increasing global prevalence. In recent years, a strong association between CP and metabolic bone diseases (MBDs), especially osteoporosis, has been identified, attracting significant attention in the research field. Epidemiological data suggest a rising trend in the incidence of MBDs among CP patients. Notably, recent studies have highlighted a profound interplay between CP and altered nutritional and immune profiles, offering insights into its linkage with MBDs. At the molecular level, CP introduces a series of biochemical disturbances that compromise bone homeostasis. One critical observation is the disrupted metabolism of vitamin D and vitamin K, both essential micronutrients for maintaining bone integrity, in CP patients. In this review, we provide physio-pathological perspectives on the development and mechanisms of CP-related MBDs. We also outline some of the latest therapeutic strategies for treating patients with CP-associated MBDs, including stem cell transplantation, monoclonal antibodies, and probiotic therapy. In summary, CP-associated MBDs represent a rising medical challenge, involving multiple tissues and organs, complex disease mechanisms, and diverse treatment approaches. More in-depth studies are required to understand the complex interplay between CP and MBDs to facilitate the development of more specific and effective therapeutic approaches.
Collapse
Affiliation(s)
- Tianlin Wang
- Department of Emergency, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ke Xiong
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanli He
- Department of General Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Binbin Feng
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - LinBin Guo
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingliang Gu
- Department of Orthopedics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengrui Zhang
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, California, United States
- Division of Immunology and Rheumatology, Stanford University, Stanford, California, United States
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States
| | - Hong Wang
- Department of General Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaohao Wu
- Division of Immunology and Rheumatology, Stanford University, Stanford, California, United States
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States
| |
Collapse
|
6
|
Ma HD, Shi L, Li HT, Wang XD, Yang MW. Polycytosine RNA-binding protein 1 regulates osteoblast function via a ferroptosis pathway in type 2 diabetic osteoporosis. World J Diabetes 2024; 15:977-987. [PMID: 38766437 PMCID: PMC11099367 DOI: 10.4239/wjd.v15.i5.977] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/22/2024] [Accepted: 03/15/2024] [Indexed: 05/10/2024] Open
Abstract
BACKGROUND Recently, type 2 diabetic osteoporosis (T2DOP) has become a research hotspot for the complications of diabetes, but the specific mechanism of its occurrence and development remains unknown. Ferroptosis caused by iron overload is con-sidered an important cause of T2DOP. Polycytosine RNA-binding protein 1 (PCBP1), an iron ion chaperone, is considered a protector of ferroptosis. AIM To investigate the existence of ferroptosis and specific role of PCBP1 in the development of type 2 diabetes. METHODS A cell counting kit-8 assay was used to detect changes in osteoblast viability under high glucose (HG) and/or ferroptosis inhibitors at different concentrations and times. Transmission electron microscopy was used to examine the morphological changes in the mitochondria of osteoblasts under HG, and western blotting was used to detect the expression levels of PCBP1, ferritin, and the ferroptosis-related protein glutathione peroxidase 4 (GPX4). A lentivirus silenced and overexpressed PCBP1. Western blotting was used to detect the expression levels of the osteoblast functional proteins osteoprotegerin (OPG) and osteocalcin (OCN), whereas flow cytometry was used to detect changes in reactive oxygen species (ROS) levels in each group. RESULTS Under HG, the viability of osteoblasts was considerably decreased, the number of mitochondria undergoing atrophy was considerably increased, PCBP1 and ferritin expression levels were increased, and GPX4 expression was decreased. Western blotting results demonstrated that infection with lentivirus overexpressing PCBP1, increased the expression levels of ferritin, GPX4, OPG, and OCN, compared with the HG group. Flow cytometry results showed a reduction in ROS, and an opposite result was obtained after silencing PCBP1. CONCLUSION PCBP1 may protect osteoblasts and reduce the harm caused by ferroptosis by promoting ferritin expression under a HG environment. Moreover, PCBP1 may be a potential therapeutic target for T2DOP.
Collapse
Affiliation(s)
- Hong-Dong Ma
- Department of Orthopedics, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Lei Shi
- Department of Orthopedics, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Hai-Tian Li
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Xin-Dong Wang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Mao-Wei Yang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
7
|
Zhang W, Rong H, Liang J, Mao C, Li Z, Dai Z, Li D, Guo W, Chen S, Wang Z, Wei J. Chitosan modified with PAP as a promising delivery system for melatonin in the treatment of osteoporosis: targeting the divalent metal transporter 1. J Biol Eng 2024; 18:27. [PMID: 38622739 PMCID: PMC11020623 DOI: 10.1186/s13036-024-00422-7] [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: 01/09/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
The demands for novel and efficient therapies have gradually increased with the rising concerns of osteoporosis (OP). The most popular method in promoting bone regeneration during osteoporotic conditions consists of loading bioactive materials with different drugs to treat osteoporotic bones by either promoting the process of osteogenesis, or by inhibiting the activity of osteoclasts. By analyzing single cell sequencing results, we found that divalent metal transporter 1 (DMT1) played a role in OP. Based on our previous results, we found that melatonin (MT) suppressed expression of DMT1 induced by high glucose during OP, so we determined the efficacy of MT for the treatment of OP. However, the clinical effects of MT on OP were unsatisfactory. To enhance its biological efficacy, we combined MT with porous gelatin chitosan (chitosan) and the conductive material, PLA-b-AP-b-PLA (PAP), then determined how MT incorporation in chitosan@PAP nanoparticles affected the ability to promote MC3T3-E1 osteogenesis and mineralization, both in vitro and in vivo. The results confirmed the effect of MT on DMT1. We then prepared and characterized composites prepared as nanofibers, and determined the efficacy of MT combined with chitosan-PAP modified hydrogels as a slow-release system in a femur model of osteoporosis mice, with associated properties suitable for bone tissue engineering. The results indicated that MT-loaded chitosan@PAP nanospheres showed favorable osteogenic functions, both in vivo and in vitro, providing a practical solution for bone regeneration for OP patients.
Collapse
Affiliation(s)
- Weilin Zhang
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Hongrui Rong
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jinguo Liang
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Chao Mao
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Zhencong Li
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Zhiwen Dai
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Dingbin Li
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Weixiong Guo
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Siyuan Chen
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Zhongwei Wang
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Jinsong Wei
- Department of Spinal Degeneration and Deformity Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
8
|
Liu LL, Liu ZR, Xiao YS, Xiao JH, Huang WM, Liu WY, Zhao K, Ye YJ. SPI1 exacerbates iron accumulation and promotes osteoclast formation through inhibiting the expression of Hepcidin. Mol Cell Endocrinol 2024; 580:112103. [PMID: 38450475 DOI: 10.1016/j.mce.2023.112103] [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: 08/01/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 03/08/2024]
Abstract
BACKGROUND Osteoporosis (OP) can be caused by an overactive osteoclastic function. Anti-osteoporosis considerable therapeutic effects in tissue repair and regeneration because bone resorption is a unique osteoclast function. In this study, we mainly explored the underlying mechanisms of osteoclasts' effects on osteoporosis. METHODS RAW264.7 cells were used and induced toward osteoclast and iron accumulation by M-CSF and RANKL administration. We investigated Hepcidin and divalent metal transporter 1 (DMT1) on iron accumulation and osteoclast formation in an ovariectomy (OVX)-induced osteoporosis. Osteoporosis was induced in mice by OVX, and treated with Hepcidin (10, 20, 40, 80 mg/kg, respectively) and overexpression of DMT1 by tail vein injection. Hepcidin, SPI1, and DMT1 were detected by immunohistochemical staining, western blot and RT-PCR. The bioinformatics assays, luciferase assays, and Chromatin Immunoprecipitation (ChIP) verified that Hepcidin was a direct SPI1 transcriptional target. Iron accumulation was detected by laser scanning confocal microscopy, Perl's iron staining and iron content assay. The formation of osteoclasts was assessed using tartrate-resistant acid phosphatase (TRAP) staining. RESULTS We found that RAW264.7 cells differentiated into osteoclasts when exposed to M-CSF and RANKL, which increased the protein levels of osteoclastogenesis-related genes, including c-Fos, MMP9, and Acp5. We also observed higher concentration of iron accumulation when M-CSF and RANKL were administered. However, Hepcidin inhibited the osteoclast differentiation cells and decreased intracellular iron concentration primary osteoclasts derived from RAW264.7. Spi-1 proto-oncogene (SPI1) transcriptionally repressed the expression of Hepcidin, increased DMT1, facilitated the differentiation and iron accumulation of mouse osteoclasts. Overexpression of SPI1 significantly declined luciferase activity of HAMP promoter and increased the enrichment of HAMP promoter. Furthermore, our results showed that Hepcidin inhibited osteoclast differentiation and iron accumulation in mouse osteoclasts and OVX mice. CONCLUSION Therefore, the study revealed that SPI1 could inhibit Hepcidin expression contribute to iron accumulation and osteoclast formation via DMT1 signaling activation in mouse with OVX.
Collapse
Affiliation(s)
- Lu-Lin Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, China
| | - Zhong-Rui Liu
- The First Clinical Medical College of Gannan Medical University, China
| | - Yao-Sheng Xiao
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, China
| | - Jian-Hua Xiao
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, China
| | - Wei-Min Huang
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, China
| | - Wu-Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, China
| | - Kai Zhao
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, China.
| | - Yong-Jun Ye
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, China.
| |
Collapse
|
9
|
Pourhanifeh MH, Hosseinzadeh A, Koosha F, Reiter RJ, Mehrzadi S. Therapeutic Effects of Melatonin in the Regulation of Ferroptosis: A Review of Current Evidence. Curr Drug Targets 2024; 25:543-557. [PMID: 38706348 DOI: 10.2174/0113894501284110240426074746] [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: 11/28/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 05/07/2024]
Abstract
Ferroptosis is implicated in the pathogenesis of multiple diseases, including neurodegenerative diseases, cardiovascular diseases, kidney pathologies, ischemia-reperfusion injury, and cancer. The current review article highlights the involvement of ferroptosis in traumatic brain injury, acute kidney damage, ethanol-induced liver injury, and PM2.5-induced lung injury. Melatonin, a molecule produced by the pineal gland and many other organs, is well known for its anti- aging, anti-inflammatory, and anticancer properties and is used in the treatment of different diseases. Melatonin's ability to activate anti-ferroptosis pathways including sirtuin (SIRT)6/p- nuclear factor erythroid 2-related factor 2 (Nrf2), Nrf2/ antioxidant responsive element (ARE)/ heme oxygenase (HO-1)/SLC7A11/glutathione peroxidase (GPX4)/ prostaglandin-endoperoxide synthase 2 (PTGS2), extracellular signal-regulated kinase (ERK)/Nrf2, ferroportin (FPN), Hippo/ Yes-associated protein (YAP), Phosphoinositide 3-kinase (PI3K)/ protein kinase B (AKT)/ mammalian target of rapamycin (mTOR) and SIRT6/ nuclear receptor coactivator 4 (NCOA4)/ ferritin heavy chain 1 (FTH1) signaling pathways suggests that it could serve as a valuable therapeutic agent for preventing cell death associated with ferroptosis in various diseases. Further research is needed to fully understand the precise mechanisms by which melatonin regulates ferroptosis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Koosha
- Department of Radiology Technology, Faculty of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cellular & Structural Biology, University of Texas, Health Science Center, San Antonio, USA
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
安 可, 周 学. [Latest Findings on Ferroptosis and Osteoarthropathy]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:1294-1299. [PMID: 38162082 PMCID: PMC10752773 DOI: 10.12182/20231160209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Indexed: 01/03/2024]
Abstract
Ferroptosis, a newly-discovered mode of programmed cell death, is closely associated with the development of various diseases throughout the human body, such as tumors of the digestive system, ischemia-reperfusion injury, osteoarthropathy, etc. Therefore, ferroptosis has become a hot research topic in many fields of study in recent years, providing new ideas for the prevention and treatment of relevant diseases. Among them, structural lesions in osteoarthropathies involving articular cartilage, subchondral bone, and synovial tissue have been found to be associated with iron overload, as well as oxidative stress, which suggests that inhibition of ferroptosis in relevant joint tissue cells may have a positive effect in halting the development of osteoarthropathy. Herein, focusing on ferroptosis and osteoarthropathy, we summarized the research developments in mechanisms related to iron metabolism and ferroptosis, analyzed the impact of ferroptosis on the pathogenesis and development of osteoarthropathy, and proposed new ideas for medication therapies of osteoarthropathy, taking into account the latest research findings.
Collapse
Affiliation(s)
- 可 安
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓病科 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 学东 周
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓病科 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Bao J, Yan Y, Zuo D, Zhuo Z, Sun T, Lin H, Han Z, Zhao Z, Yu H. Iron metabolism and ferroptosis in diabetic bone loss: from mechanism to therapy. Front Nutr 2023; 10:1178573. [PMID: 37215218 PMCID: PMC10196368 DOI: 10.3389/fnut.2023.1178573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/07/2023] [Indexed: 05/24/2023] Open
Abstract
Osteoporosis, one of the most serious and common complications of diabetes, has affected the quality of life of a large number of people in recent years. Although there are many studies on the mechanism of diabetic osteoporosis, the information is still limited and there is no consensus. Recently, researchers have proven that osteoporosis induced by diabetes mellitus may be connected to an abnormal iron metabolism and ferroptosis inside cells under high glucose situations. However, there are no comprehensive reviews reported. Understanding these mechanisms has important implications for the development and treatment of diabetic osteoporosis. Therefore, this review elaborates on the changes in bones under high glucose conditions, the consequences of an elevated glucose microenvironment on the associated cells, the impact of high glucose conditions on the iron metabolism of the associated cells, and the signaling pathways of the cells that may contribute to diabetic bone loss in the presence of an abnormal iron metabolism. Lastly, we also elucidate and discuss the therapeutic targets of diabetic bone loss with relevant medications which provides some inspiration for its cure.
Collapse
Affiliation(s)
- Jiahao Bao
- Department of Oral & Cranio-maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yixuan Yan
- Guangdong Provincial Key Laboratory of Stomatology, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Daihui Zuo
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Zhiyong Zhuo
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Tianhao Sun
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Hongli Lin
- School of Public Health, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Zheshen Han
- School of Public Health, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Zhiyang Zhao
- Department of Oral & Cranio-maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Hongbo Yu
- Department of Oral & Cranio-maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
12
|
Fighting age-related orthopedic diseases: focusing on ferroptosis. Bone Res 2023; 11:12. [PMID: 36854703 PMCID: PMC9975200 DOI: 10.1038/s41413-023-00247-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/03/2023] [Accepted: 01/11/2023] [Indexed: 03/02/2023] Open
Abstract
Ferroptosis, a unique type of cell death, is characterized by iron-dependent accumulation and lipid peroxidation. It is closely related to multiple biological processes, including iron metabolism, polyunsaturated fatty acid metabolism, and the biosynthesis of compounds with antioxidant activities, including glutathione. In the past 10 years, increasing evidence has indicated a potentially strong relationship between ferroptosis and the onset and progression of age-related orthopedic diseases, such as osteoporosis and osteoarthritis. Therefore, in-depth knowledge of the regulatory mechanisms of ferroptosis in age-related orthopedic diseases may help improve disease treatment and prevention. This review provides an overview of recent research on ferroptosis and its influences on bone and cartilage homeostasis. It begins with a brief overview of systemic iron metabolism and ferroptosis, particularly the potential mechanisms of ferroptosis. It presents a discussion on the role of ferroptosis in age-related orthopedic diseases, including promotion of bone loss and cartilage degradation and the inhibition of osteogenesis. Finally, it focuses on the future of targeting ferroptosis to treat age-related orthopedic diseases with the intention of inspiring further clinical research and the development of therapeutic strategies.
Collapse
|
13
|
Gao L, Hua W, Tian L, Zhou X, Wang D, Yang Y, Ni G. Molecular Mechanism of Ferroptosis in Orthopedic Diseases. Cells 2022; 11:2979. [PMID: 36230941 PMCID: PMC9563396 DOI: 10.3390/cells11192979] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 01/17/2023] Open
Abstract
Ferroptosis is a new iron-dependent programmed cell death process that is directly mediated by the accumulation of lipid peroxides and reactive oxygen species. Numerous studies have shown that ferroptosis is important in regulating the occurrence and development of bone-related diseases, but the underlying mechanisms are not completely clear. Herein, we review the progress of the mechanism of ferroptosis in bone marrow injury, osteoporosis, osteoarthritis, and osteosarcoma and attempt to deeply understand the regulatory targets of ferroptosis, which will open up a new way for the prevention and treatment of orthopedic diseases.
Collapse
Affiliation(s)
- Lu Gao
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Weizhong Hua
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Lixiang Tian
- School of Physical Education, Shanghai University, Shanghai 200444, China
| | - Xuchang Zhou
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Dongxue Wang
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Yajing Yang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Guoxin Ni
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| |
Collapse
|
14
|
Ren Y, Yang M, Wang X, Xu B, Xu Z, Su B. ELAV-like RNA binding protein 1 regulates osteogenesis in diabetic osteoporosis: Involvement of divalent metal transporter 1. Mol Cell Endocrinol 2022; 546:111559. [PMID: 35051552 DOI: 10.1016/j.mce.2022.111559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/23/2021] [Accepted: 01/10/2022] [Indexed: 12/18/2022]
Abstract
Diabetic osteoporosis (DOP) is a complication of diabetes mellitus (DM) and occurs due to alterations in bone metabolism under hyperglycemic condition. ELAV-like RNA binding protein 1 (ELAVL1) is abnormally up-regulated in diabetes-related diseases. Bioinformatics prediction indicates that divalent metal transporter 1 (DMT1) is a potential target of ELAVL1. To explore the role of ELAVL1 and the involvement of ELAVL1/DMT1 axis in DOP, we established a mouse model of DM by administration of high-fat diet and intraperitoneal injection with streptozotocin (STZ). The expression of ELAVL1 and DMT1 was increased in the bone tissues of DM mice. Knockdown of ELAVL1 reduced iron level and oxidative stress, promoted osteogensis, and prevented bone mass loss, thus mitigating DOP in DM mice. In vitro, mouse pre-osteoblast MC3T3-E1 cells were treated with high glucose (25 mM) and ferric ammonium citrate (FAC, 200 μM). The inhibitory effects of ELAVL1 knockdown on iron accumulation and oxidative stress were evidenced in MC3T3-E1 cells. Knockdown of ELAVL1 enhanced osteoblast viability, differentiation and mineralization. Notably, the expression of DMT1 was positively correlated with that of ELAVL1 in vivo and in vitro. Overexpression of DMT1 abolished the effect of ELAVL1 knockdown on the behaviors of MC3T3-E1 cells, suggesting that ELAVL1 might function through regulating DMT1. In conclusion, knockdown of ELAVL1 likely alleviated DOP by inhibiting iron overload and oxidative stress and promoting osteogenesis, and DMT1 might be involved in this process. These findings provide insights into the pathogenesis of DOP and suggest a potential therapeutic target for DOP treatment.
Collapse
Affiliation(s)
- Yuanfei Ren
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China; The First Department of Hand and Foot Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China
| | - Maowei Yang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Xindong Wang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Buxuan Xu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zerong Xu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bo Su
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
15
|
Lin Y, Shen X, Ke Y, Lan C, Chen X, Liang B, Zhang Y, Yan S. Activation of osteoblast ferroptosis via the METTL3/ASK1‐p38 signaling pathway in high glucose and high fat (HGHF)‐induced diabetic bone loss. FASEB J 2022; 36:e22147. [PMID: 35104016 DOI: 10.1096/fj.202101610r] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/11/2021] [Accepted: 12/21/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Youfen Lin
- Department of Endocrinology the First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Ximei Shen
- Department of Endocrinology the First Affiliated Hospital of Fujian Medical University Fuzhou China
- Clinical Research Center for Metabolic Diseases of Fujian Province the First Affiliated Hospital of Fujian Medical University Fuzhou China
- Diabetes Research Institute of Fujian Province the First Affiliated Hospital of Fujian Medical University Fuzhou China
- Metabolic Diseases Research Institute the First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Yuzhen Ke
- Department of Endocrinology the First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Chao Lan
- Department of Endocrinology the First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Xiaoyuan Chen
- Department of Endocrinology the First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Bo Liang
- Department of Endocrinology the First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Yongze Zhang
- Department of Endocrinology the First Affiliated Hospital of Fujian Medical University Fuzhou China
| | - Sunjie Yan
- Department of Endocrinology the First Affiliated Hospital of Fujian Medical University Fuzhou China
- Clinical Research Center for Metabolic Diseases of Fujian Province the First Affiliated Hospital of Fujian Medical University Fuzhou China
- Diabetes Research Institute of Fujian Province the First Affiliated Hospital of Fujian Medical University Fuzhou China
- Metabolic Diseases Research Institute the First Affiliated Hospital of Fujian Medical University Fuzhou China
| |
Collapse
|
16
|
Cai C, Hu W, Chu T. Interplay Between Iron Overload and Osteoarthritis: Clinical Significance and Cellular Mechanisms. Front Cell Dev Biol 2022; 9:817104. [PMID: 35096841 PMCID: PMC8795893 DOI: 10.3389/fcell.2021.817104] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/28/2021] [Indexed: 01/15/2023] Open
Abstract
There are multiple diseases or conditions such as hereditary hemochromatosis, hemophilia, thalassemia, sickle cell disease, aging, and estrogen deficiency that can cause iron overload in the human body. These diseases or conditions are frequently associated with osteoarthritic phenotypes, such as progressive cartilage degradation, alterations in the microarchitecture and biomechanics of the subchondral bone, persistent joint inflammation, proliferative synovitis, and synovial pannus. Growing evidences suggest that the conditions of pathological iron overload are associated with these osteoarthritic phenotypes. Osteoarthritis (OA) is an important complication in patients suffering from iron overload-related diseases and conditions. This review aims to summarize the findings and observations made in the field of iron overload-related OA while conducting clinical and basic research works. OA is a whole-joint disease that affects the articular cartilage lining surfaces of bones, subchondral bones, and synovial tissues in the joint cavity. Chondrocytes, osteoclasts, osteoblasts, and synovial-derived cells are involved in the disease. In this review, we will elucidate the cellular and molecular mechanisms associated with iron overload and the negative influence that iron overload has on joint homeostasis. The promising value of interrupting the pathologic effects of iron overload is also well discussed for the development of improved therapeutics that can be used in the field of OA.
Collapse
Affiliation(s)
- Chenhui Cai
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tongwei Chu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
17
|
Wang X, Ma H, Sun J, Zheng T, Zhao P, Li H, Yang M. Mitochondrial Ferritin Deficiency Promotes Osteoblastic Ferroptosis Via Mitophagy in Type 2 Diabetic Osteoporosis. Biol Trace Elem Res 2022; 200:298-307. [PMID: 33594527 DOI: 10.1007/s12011-021-02627-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/03/2021] [Indexed: 12/15/2022]
Abstract
The incidence of type 2 diabetic osteoporosis (T2DOP), which seriously threatens elderly people's health, is rapidly increasing in recent years. However, the specific mechanism of the T2DOP is still unclear. Studies have shown the relationship between iron overload and T2DOP. Mitochondrial ferritin (FtMt) is a protein that stores iron ions and intercepts toxic ferrous ions in cells mitochondria. Ferroptosis, an iron-dependent cell injured way, may be related to the pathogenesis of T2DOP. In this study, we intend to elucidate the effect of FtMt on ferroptosis in osteoblasts and explain the possible mechanism. We first detected the occurrence of ferroptosis in bone tissue and the expression of FtMt after inducing T2DOP rat model. Then we used hFOB1.19 cells to study the influence of high glucose on FtMt, ferroptosis, and osteogenic function of osteoblasts. Then we observed the effect of FtMt on ferroptosis and osteoblast function by lentiviral silencing and overexpression of FtMt. We found ferroptosis in T2DOP rats bone. Overexpression of FtMt reduced osteoblastic ferroptosis under high glucose condition while silent FtMt induced mitophagy through ROS / PINK1/Parkin pathway. Then we found increased ferroptosis in osteoblasts after activating mitophagy by carbonyl cyanide-m-chlorophenyl-hydrazine (CCCP, a mitophagy agonist). Our study demonstrated that FtMt inhibited the occurrence of ferroptosis in osteoblasts by reducing oxidative stress caused by excess ferrous ions, and FtMt deficiency induced mitophagy in the pathogenesis of T2DOP. This study suggested that FtMt might serve as a potential target for T2DOP therapy.
Collapse
Affiliation(s)
- XinDong Wang
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - HongDong Ma
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Jun Sun
- Department of Orthopedics, The Third Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - TianYu Zheng
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, China
| | - Peng Zhao
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - HaiTian Li
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - MaoWei Yang
- Department of Orthopedics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
18
|
Sun K, Guo Z, Hou L, Xu J, Du T, Xu T, Guo F. Iron homeostasis in arthropathies: From pathogenesis to therapeutic potential. Ageing Res Rev 2021; 72:101481. [PMID: 34606985 DOI: 10.1016/j.arr.2021.101481] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/25/2021] [Accepted: 09/29/2021] [Indexed: 02/08/2023]
Abstract
Iron is an essential element for proper functioning of cells within mammalian organ systems; in particular, iron homeostasis is critical for joint health. Excess iron can induce oxidative stress damage, associated with the pathogenesis of iron-storage and ageing-related diseases. Therefore, iron levels in body tissues and cells must be tightly regulated. In the past decades, excess iron content within joints has been found in some patients with joint diseases including hemophilic arthropathy, hemochromatosis arthropathy, and osteoarthritis (OA). Currently, increased evidence has shown that iron accumulation is closely associated with multiple pathological changes of these arthropathies. This review summarizes system-level and intracellular regulation of iron homeostasis, and emphasizes the role of iron in synovial alterations, cartilage degeneration, and subchondral bone of several arthropathies. Of note, we discuss the potential link between iron homeostasis and OA pathogenesis. Finally, we discuss the therapeutic potential of maintaining iron homeostasis in these arthropathies.
Collapse
|
19
|
Shang M, Weng L, Xu G, Wu S, Liu B, Yin X, Mao A, Zou X, Wang Z. TRIM11 suppresses ferritinophagy and gemcitabine sensitivity through UBE2N/TAX1BP1 signaling in pancreatic ductal adenocarcinoma. J Cell Physiol 2021; 236:6868-6883. [PMID: 33629745 DOI: 10.1002/jcp.30346] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 02/01/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022]
Abstract
Gemcitabine is first-line chemotherapy for pancreatic cancer, however, the development of resistance limits its effectiveness. The tripartite motif-containing 11 (TRIM11) protein plays crucial roles in tumor development and undergoes auto-polyubiquitination to promote interactions in selective autophagy. Therefore, Understanding whether TRIM11 is involved in ferritinophagy and gemcitabine resistance in pancreatic cancer is critical in developing pancreatic cancer therapeutics. TRIM11 expression was validated by Western blot analysis, real-time polymease chain reaction, and immunohistochemical staining. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and Colony formation assays were performed to investigate pancreatic ductal adenocarcinomas (PDAC) cell viability. Mouse xenograft model of PDAC cells was established to verify the role of TRIM11 in vivo. Coimmunoprecipitation was used to identify the reciprocal regulation between TRIM11 and UBE2N. In this study, we found that TRIM11 expression were higher in PDAC cells and tissues. TRIM11 overexpression promotes PDAC cell proliferation in vitro and tumor growth in vivo. Decreased expression of TRIM11 in PDAC patients is associated with decreased UBE2N and increased TAX1BP1 expression. Coimmunoprecipitation established that TRIM11 interacts and colocalizes with UBE2N. Mechanistically, TRIM11 promoted gemcitabine resistance and suppressed ferritinophagy through UBE2N-TAX1BP1 signaling. Our findings identify TRIM11 as a key regulator of TAX1BP1 signaling with a crucial role in ferritinophagy and gemcitabine resistance in PDAC.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Autophagy/drug effects
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Drug Resistance, Neoplasm
- Female
- Ferroptosis/drug effects
- Gene Expression Regulation, Neoplastic
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Signal Transduction
- Tripartite Motif Proteins/genetics
- Tripartite Motif Proteins/metabolism
- Tumor Burden/drug effects
- Ubiquitin-Conjugating Enzymes/genetics
- Ubiquitin-Conjugating Enzymes/metabolism
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
- Xenograft Model Antitumor Assays
- Gemcitabine
- Mice
Collapse
Affiliation(s)
- Mingyi Shang
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Li Weng
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guifang Xu
- Department of gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Shaoqiu Wu
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bingyan Liu
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiang Yin
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Aiwu Mao
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoping Zou
- Department of interventional radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongmin Wang
- Department of gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
20
|
Divalent Metal Transporter 1 Knock-Down Modulates IL-1β Mediated Pancreatic Beta-Cell Pro-Apoptotic Signaling Pathways through the Autophagic Machinery. Int J Mol Sci 2021; 22:ijms22158013. [PMID: 34360779 PMCID: PMC8348373 DOI: 10.3390/ijms22158013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/21/2022] Open
Abstract
Pro-inflammatory cytokines promote cellular iron-import through enhanced divalent metal transporter-1 (DMT1) expression in pancreatic β-cells, consequently cell death. Inhibition of β-cell iron-import by DMT1 silencing protects against apoptosis in animal models of diabetes. However, how alterations of signaling networks contribute to the protective action of DMT1 knock-down is unknown. Here, we performed phosphoproteomics using our sequential enrichment strategy of mRNA, protein, and phosphopeptides, which enabled us to explore the concurrent molecular events in the same set of wildtype and DMT1-silenced β-cells during IL-1β exposure. Our findings reveal new phosphosites in the IL-1β-induced proteins that are clearly reverted by DMT1 silencing towards their steady-state levels. We validated the levels of five novel phosphosites of the potential protective proteins using parallel reaction monitoring. We also confirmed the inactivation of autophagic flux that may be relevant for cell survival induced by DMT1 silencing during IL-1β exposure. Additionally, the potential protective proteins induced by DMT1 silencing were related to insulin secretion that may lead to improving β-cell functions upon exposure to IL-1β. This global profiling has shed light on the signal transduction pathways driving the protection against inflammation-induced cell death in β-cells after DMT1 silencing.
Collapse
|
21
|
Ma H, Wang X, Zhang W, Li H, Zhao W, Sun J, Yang M. Melatonin Suppresses Ferroptosis Induced by High Glucose via Activation of the Nrf2/HO-1 Signaling Pathway in Type 2 Diabetic Osteoporosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9067610. [PMID: 33343809 PMCID: PMC7732386 DOI: 10.1155/2020/9067610] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/18/2020] [Accepted: 11/18/2020] [Indexed: 12/25/2022]
Abstract
Ferroptosis is recently identified, an iron- and reactive oxygen species- (ROS-) dependent form of regulated cell death. This study was designed to determine the existence of ferroptosis in the pathogenesis of type 2 diabetic osteoporosis and confirm that melatonin can inhibit the ferroptosis of osteoblasts through activating Nrf2/HO-1 signaling pathway to improve bone microstructure in vivo and in vitro. We treated MC3T3-E1 cells with different concentrations of melatonin (1, 10, or 100 μM) and exposed them to high glucose (25.5 mM) for 48 h in vitro. Our data showed that high glucose can induce osteoblast cytotoxicity and the accumulation of lipid peroxide, the mitochondria of osteoblast show the same morphology changes as the erastin treatment group, and the expression of ferroptosis-related proteins glutathione peroxidase 4 (GPX4) and cystine-glutamate antiporter (SLC7A11) is downregulated, but these effects were reversed by ferroptosis inhibitor ferrastatin-1 and iron chelator deferoxamine (DFO). Furthermore, western blot and real-time polymerase chain reaction were used to detect the expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1); osteogenic capacity was evaluated by alizarin red S staining and the expression of osteoprotegerin, osteocalcin, and alkaline phosphatase; the results showed that the expression levels of these proteins in osteoblasts with 1, 10, or 100 μM melatonins were significantly higher than the high glucose group, but after using Nrf2-SiRNA interference, the therapeutic effect of melatonin was significantly inhibited. We also performed in vivo experiments in a diabetic rat model treated with two concentrations of melatonin (10, 50 mg/kg). Dynamic bone histomorphometry and micro-CT were used to observe the rat bone microstructure, and the expression of GPX4 and Nrf2 was determined by immunohistochemistry. Here, we first report that high glucose induces ferroptosis via increased ROS/lipid peroxidation/glutathione depletion in type 2 diabetic osteoporosis. More importantly, melatonin significantly reduced the level of ferroptosis and improved the osteogenic capacity of MC3T3-E1 through activating the Nrf2/HO-1 pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Hongdong Ma
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xindong Wang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Weilin Zhang
- Department of Orthopedics, The Fourth Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haitian Li
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wei Zhao
- Department of Orthopedics, The Fourth Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jun Sun
- Department of Orthopedics, The Third Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Maowei Yang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
22
|
Zhang S, Sun L, Zhang J, Liu S, Han J, Liu Y. Adverse Impact of Heavy Metals on Bone Cells and Bone Metabolism Dependently and Independently through Anemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000383. [PMID: 33042736 PMCID: PMC7539179 DOI: 10.1002/advs.202000383] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/21/2020] [Indexed: 05/05/2023]
Abstract
Mounting evidence is revealing that heavy metals can incur disordered bone homeostasis, leading to the development of degenerative bone diseases, including osteoporosis, osteoarthritis, degenerative disk disease, and osteomalacia. Meanwhile, heavy metal-induced anemia has been found to be intertwined with degenerative bone diseases. However, the relationship and interplay among these adverse outcomes remain elusive. Thus, it is of importance to shed light on the modes of action (MOAs) and adverse outcome pathways (AOPs) responsible for degenerative bone diseases and anemia under exposure to heavy metals. In the current Review, the epidemiological and experimental findings are recapitulated to interrogate the contributions of heavy metals to degenerative bone disease development which may be attributable dependently and independently to anemia. A few likely mechanisms are postulated for anemia-independent degenerative bone diseases, including dysregulated osteogenesis and osteoblastogenesis, imbalanced bone formation and resorption, and disturbed homeostasis of essential trace elements. By contrast, remodeled bone microarchitecture, inhibited erythropoietin production, and disordered iron homeostasis are speculated to account for anemia-associated degenerative bone disorders upon heavy metal exposure. Together, this Review aims to elaborate available literature to fill in the knowledge gaps in understanding the detrimental effects of heavy metals on bone cells and bone homeostasis through different perspectives.
Collapse
Affiliation(s)
- Shuping Zhang
- The First Affiliated Hospital of Shandong First Medical UniversityJinanShandong250014China
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250062China
| | - Li Sun
- The First Affiliated Hospital of Shandong First Medical UniversityJinanShandong250014China
| | - Jie Zhang
- The First Affiliated Hospital of Shandong First Medical UniversityJinanShandong250014China
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250062China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085China
| | - Jinxiang Han
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250062China
| | - Yajun Liu
- Beijing Jishuitan HospitalPeking University Health Science CenterBeijing100035China
| |
Collapse
|
23
|
Abstract
INTRODUCTION Iron overload, a state with excessive iron storage in the body, is a common complication in thalassemia patients which leads to multiple organ dysfunctions including the bone. Iron overload-induced bone disease is one of the most common and severe complications of thalassemia including osteoporosis. Currently, osteoporosis is still frequently found in thalassemia even with widely available iron chelation therapy. STUDY SELECTION Relevant publications published before December 2019 in PubMed database were reviewed. Both pre-clinical studies and clinical trials were obtained using iron overload, thalassemia, osteoporosis, osteoblast, and osteoclast as keywords. RESULTS Increased ROS production is a hallmark of iron overload-induced impaired bone remodeling. At the cellular level, oxidative stress affects bone remodeling by both osteoblast inhibition and osteoclast activation via many signaling pathways. In thalassemia patients, it has been shown that bone resorption was increased while bone formation was concurrently reduced. CONCLUSION In this review, reports on the cellular mechanisms of iron overload-associated bone remodeling are comprehensively summarized and presented to provide current understanding this pathological condition. Moreover, current treatments and potential interventions for attenuating bone remodeling in iron overload are also summarized to pave ways for the future discoveries of novel agents that alleviate this condition.
Collapse
|
24
|
Zhu X, Zhao Z, Zeng C, Chen B, Huang H, Chen Y, Zhou Q, Yang L, Lv J, Zhang J, Pan D, Shen J, Duque G, Cai D. HNGF6A Inhibits Oxidative Stress-Induced MC3T3-E1 Cell Apoptosis and Osteoblast Phenotype Inhibition by Targeting Circ_0001843/miR-214 Pathway. Calcif Tissue Int 2020; 106:518-532. [PMID: 32189040 DOI: 10.1007/s00223-020-00660-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 01/11/2020] [Indexed: 01/08/2023]
Abstract
Humanin (HN), a mitochondrial derived peptide, plays cyto-protective role under various stress. In this study, we aimed to investigate the effects of HNGF6A, an analogue of HN, on osteoblast apoptosis and differentiation and the underlying mechanisms. Cell proliferation of murine osteoblastic cell line MC3TC-E1 was examined by CCK8 assay and Edu staining. Cell apoptosis was detected by Annexin V assay under H2O2 treatment. The differentiation of osteoblast was determined by Alizarin red S staining. We also tested the expression of osteoblast phenotype related protein by real-time PCR and Western blot. The interaction between Circ_0001843 and miR-214, miR-214 and TAFA5 was examined by luciferase report assay. Circ_0001843 was inhibited by siRNA and miR-214 was suppressed by miR-214 inhibitor to determine the effects of Circ_0001843 and miR-214 on cell proliferation, apoptosis, and differentiation. HNGF6A, an analogue of HN, exerted cyto-protection and osteogenesis-promotion in MC3T3-E1 cells. The expression of osteoblast phenotype related protein was significantly induced by HNGF6A. Additionally, HNGF6A treatment decreased Circ_0001843 and increased miR-214 levels, as well as inhibited the phosphorylation of p38 and JNK. We further found that Circ_0001843 directly bound with miR-214, which in turn inhibited the phosphorylation of p38 and JNK. Furthermore, both Circ_0001843 overexpression and miR-214 knockdown significantly decreased the cyto-protection and osteogenic promotion of HNGF6A. In summary, our data showed that HNGF6A protected osteoblasts from oxidative stress-induced apoptosis and osteoblast phenotype inhibition by targeting Circ_0001843/miR-214 pathway and the downstream kinases, p38 and JNK.
Collapse
Affiliation(s)
- Xiao Zhu
- Department of Endocrinology, The Third Affiliated Hospital of Southern Medical University, No. 183 West Zhongshan Road, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Ziping Zhao
- Department of Joint Surgery, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, No.183 West Zhongshan Road, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Canjun Zeng
- Department of Foot and Ankle Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Bo Chen
- Department of Endocrinology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Haifeng Huang
- Department of Internal Medicine, the Eastern Hospital of the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510700, Guangdong, China
| | - Youming Chen
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Quan Zhou
- Department of Medical Image, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Li Yang
- Department of Endocrinology, People's Hospital of Hunan Province, Changsha, 410011, Hunan, China
| | - Jicheng Lv
- Department of Endocrinology, The Third Affiliated Hospital of Southern Medical University, No. 183 West Zhongshan Road, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Jing Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Daoyan Pan
- Department of Endocrinology, The Third Affiliated Hospital of Southern Medical University, No. 183 West Zhongshan Road, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Jie Shen
- Department of Endocrinology, The Third Affiliated Hospital of Southern Medical University, No. 183 West Zhongshan Road, Tianhe District, Guangzhou, 510630, Guangdong, China.
| | - Gustavo Duque
- Department of Medicine, Western Health, The University of Melbourne, St Albans, Victoria, 3021, Australia.
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St Albans, Victoria, 3021, Australia.
| | - Daozhang Cai
- Department of Joint Surgery, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, No.183 West Zhongshan Road, Tianhe District, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
25
|
Li G, Zhang H, Wu J, Wang A, Yang F, Chen B, Gao Y, Ma X, Xu Y. Hepcidin deficiency causes bone loss through interfering with the canonical Wnt/β-catenin pathway via Forkhead box O3a. J Orthop Translat 2020; 23:67-76. [PMID: 32514392 PMCID: PMC7267010 DOI: 10.1016/j.jot.2020.03.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 03/01/2020] [Accepted: 03/23/2020] [Indexed: 01/05/2023] Open
Abstract
Objective Hepcidin deficiency is known to cause body iron accumulation and bone microarchitecture defects, but the exact underlying mechanisms of hepcidin deficiency-induced bone loss remain unclear. Our objective was to understand the molecular mechanism of hepcidin deficiency-induced bone loss. Methods The bone phenotypes of wild type (WT) and hepcidin knockout (Hepcidin-KO) mice were measured by microcomputed tomography. The osteoclastic marker of the bone was measured by tartrate-resistant acid phosphatase staining. The osteoblastic marker of the bone was measured by immunostaining of osteocalcin. Primary osteoblastic and osteoclastic differentiation was performed using bone marrow cells. The mature osteoclast was determined by tartrate-resistant acid phosphatase staining, pit formation assay and relative gene expression. The mature osteoblast was determined by alkaline phosphatase activity, alkaline phosphatase staining, Alizarin Red staining and relative gene expression. The protein expression of β-catenin, TCF4/TCF7L2 and Forkhead box O3a (FOXO3a) was measured by Western blot and their combination by co-immunoprecipitation. In vivo study was performed by tail vein administration of FOXO3a-RNAi using an adeno-associated virus in Hepcidin-KO mice. Results We found that Hepcidin-KO mice exhibited iron accumulation and bone loss compared with WT mice. The osteoclastic differentiation of bone marrow-derived macrophages from Hepcidin-KO mice was not significantly different from that of bone marrow–derived macrophages from WT mice. However, the osteoblastic differentiation of bone marrow–derived mesenchymal stem cells from Hepcidin-KO mice was obviously decreased compared with that of bone marrow–derived mesenchymal stem cells from WT mice. Furthermore, it was confirmed in this study that upon hepcidin deficiency, β-catenin, TCF4/TCF7L2 and FOXO3a expression in bone tissues was not altered, but β-catenin combination with TCF4/TCF7L2 was strongly inhibited by β-catenin combination with FOXO3a, indicating that the canonical Wnt/β-catenin pathway was affected. Tail vein administration of FOXO3a-RNAi using an adeno-associated virus in Hepcidin-KO mice resulted in bone mass recovery. Conclusion These findings suggested that hepcidin deficiency might cause bone loss by interfering with the canonical Wnt/β-catenin pathway via FOXO3a, and FOXO3a inhibition would be a possible approach to treat hepcidin deficiency-induced bone loss. The translational potential of this article Hepcidin deficiency, as well as iron accumulation, has been considered as a risk factor for osteoporosis. For this kind of osteoporosis, inhibition of FOXO3a either by neutralized antibody or AAV-mediated RNAi, represents an effective and promising method.
Collapse
Affiliation(s)
- Guangfei Li
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Hui Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Jiadong Wu
- Department of Orthopaedics, The Affiliated Yancheng Hospital of Southeast University Medical College, 224005, Yancheng, China
| | - Aifei Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Fan Yang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Bin Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Yan Gao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Xiaowei Ma
- Department of Orthopaedics, Zhongshan Hospital of Dalian University, 116001, Dalian, China
| | - Youjia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| |
Collapse
|
26
|
Biological Factors, Metals, and Biomaterials Regulating Osteogenesis through Autophagy. Int J Mol Sci 2020; 21:ijms21082789. [PMID: 32316424 PMCID: PMC7215394 DOI: 10.3390/ijms21082789] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 01/18/2023] Open
Abstract
Bone loss raises great concern in numerous situations, such as ageing and many diseases and in both orthopedic and dentistry fields of application, with an extensive impact on health care. Therefore, it is crucial to understand the mechanisms and the determinants that can regulate osteogenesis and ensure bone balance. Autophagy is a well conserved lysosomal degradation pathway, which is known to be highly active during differentiation and development. This review provides a revision of the literature on all the exogen factors that can modulate osteogenesis through autophagy regulation. Metal ion exposition, mechanical stimuli, and biological factors, including hormones, nutrients, and metabolic conditions, were taken into consideration for their ability to tune osteogenic differentiation through autophagy. In addition, an exhaustive overview of biomaterials, both for orthopedic and dentistry applications, enhancing osteogenesis by modulation of the autophagic process is provided as well. Already investigated conditions regulating bone regeneration via autophagy need to be better understood for finely tailoring innovative therapeutic treatments and designing novel biomaterials.
Collapse
|
27
|
NIPA2 regulates osteoblast function by modulating mitophagy in type 2 diabetes osteoporosis. Sci Rep 2020; 10:3078. [PMID: 32080264 PMCID: PMC7033235 DOI: 10.1038/s41598-020-59743-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023] Open
Abstract
The highly selective magnesium transporter non-imprinted in Prader-Willi/Angelman syndrome region protein 2 (NIPA2) has recently been associated with the development and progression of type 2 diabetes osteoporosis, but the mechanisms involved are still poorly understood. Because mitophagy is involved in the pathology of type 2 diabetes osteoporosis, the present study aimed to explore the relationship among NIPA2, mitophagy and osteoblast osteogenic capacity. NIPA2 expression was reduced in C57BKS background db/db mice and in vitro models of type 2 diabetes osteoporosis, and the activation of mitophagy in primary culture osteoblast-derived from db/db mice and in high glucose-treated human fetal osteoblastic cells (hFOB1.19) was observed. Knockdown, overexpression of NIPA2 and pharmacological inhibition of peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) showed that NIPA2 increased osteoblast function, which was likely regulated by PTEN induced kinase 1 (PINK1)/E3 ubiquitin ligase PARK2 (Parkin)-mediated mitophagy via the PGC-1α/forkhead box O3a(FoxO3a)/mitochondrial membrane potential (MMP) pathway. Furthermore, the negative effect of mitophagy on osteoblast function was confirmed by pharmacological regulation of mitophagy and knockdown of Parkin. Taken together, these results suggest that NIPA2 positively regulates the osteogenic capacity of osteoblasts via the mitophagy pathway in type 2 diabetes.
Collapse
|
28
|
Wei S, Qiu T, Yao X, Wang N, Jiang L, Jia X, Tao Y, Wang Z, Pei P, Zhang J, Zhu Y, Yang G, Liu X, Liu S, Sun X. Arsenic induces pancreatic dysfunction and ferroptosis via mitochondrial ROS-autophagy-lysosomal pathway. JOURNAL OF HAZARDOUS MATERIALS 2020; 384:121390. [PMID: 31735470 DOI: 10.1016/j.jhazmat.2019.121390] [Citation(s) in RCA: 272] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 05/16/2023]
Abstract
Chronic arsenic exposure is a significantly risk factor for pancreatic dysfunction and type 2 diabetes (T2D). Ferroptosis is a newly identified iron-dependent form of oxidative cell death that relies on lipid peroxidation. Previous data have indicated that ferroptosis is involved in various diseases, including cancers, neurodegenerative diseases, and T2D. However, the concrete effect and mechanism of ferroptosis on pancreatic dysfunction triggered by arsenic remains unknown. In this study, we verified that ferroptosis occurred in animal models of arsenic-induced pancreatic dysfunction through assessing proferroptotic markers and morphological changes in mitochondria. In vitro, arsenic caused execution of ferroptosis in a dose-dependent manner, which could be significantly reduced by ferrostatin-1. Additionally, arsenic damaged mitochondria manifested as diminishing of mitochondrial membrane potential, reduced cytochrome c level and production of mitochondrial reactive oxygen species (MtROS) in MIN6 cells. Using the Mito-TEMPO, we found the autophagy level and subsequent ferroptotic cell death induced by arsenic were both alleviated. With autophagy inhibitor chloroquine, we further revealed that ferritin regulated ferroptosis through the MtROS-autophagy pathway. Collectively, NaAsO2-induced ferroptotic cell death is relied on the MtROS-dependent autophagy by regulating the iron homeostasis. Ferroptosis is involved in pancreatic dysfunction triggered by arsenic, and arsenic-induced ferroptosis involves MtROS, autophagy, ferritin.
Collapse
Affiliation(s)
- Sen Wei
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Tianming Qiu
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Xiaofeng Yao
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Ningning Wang
- Experimental Teaching Center of Public Health, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Liping Jiang
- Experimental Teaching Center of Public Health, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Xue Jia
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Ye Tao
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Zhidong Wang
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Pei Pei
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Jingyuan Zhang
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Yuhan Zhu
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Guang Yang
- Nutrition and Food Hygiene, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Xiaofang Liu
- Nutrition and Food Hygiene, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Shuang Liu
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Xiance Sun
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China; Global Health Research Center, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| |
Collapse
|
29
|
Zhao W, Zhang WL, Yang B, Sun J, Yang MW. NIPA2 regulates osteoblast function via its effect on apoptosis pathways in type 2 diabetes osteoporosis. Biochem Biophys Res Commun 2019; 513:883-890. [PMID: 31003774 DOI: 10.1016/j.bbrc.2019.04.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 04/03/2019] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes osteoporosis has recently become a hot topic in the study of diabetic complications, but the specific mechanism of its development remains unclear. Non-imprinted in Prader-Willi/Angelman syndrome region protein 2 (NIPA2), a highly-selective magnesium ion transporter, has been found to be associated with type 2 diabetes. In this study we aimed to investigate the specific role and mechanism of NIPA2 in the pathogenesis of type 2 diabetes osteoporosis. We first used western blotting, PCR, immunofluorescence, and magnesium ion probes to detect changes of NIPA2 and intracellular magnesium levels in osteoblasts at different concentrations of advanced glycation end products (AGEs). We then up- or down-regulated NIPA2 using a lentivirus and analyzed apoptotic biomarkers as well as the osteogenic ability of osteoblasts. We found that AGEs dose-dependently down-regulated the expression of NIPA2 in osteoblasts. NIPA2 also regulated osteoblast apoptosis by affecting the intracellular magnesium level and further affecting the osteogenic capacity of osteoblasts. Our study revealed the changes of NIPA2 in response to AGEs in the environment, as well as its function and mechanism in osteoblasts, demonstrating its important role in the pathogenesis of type 2 diabetes osteoporosis. The study suggests that NIPA2 is a potential target for the treatment of type 2 diabetes osteoporosis.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wei-Lin Zhang
- Department of Orthopedics, The Fourth Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bo Yang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jun Sun
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mao-Wei Yang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
30
|
Paisrisarn P, Tepaamorndech S, Khongkow M, Khemthong P, Kasamechonchung P, Klysubun W, Wutikhun T, Huang L, Chantarasakha K, Boonrungsiman S. Alterations of mineralized matrix by lead exposure in osteoblast (MC3T3-E1) culture. Toxicol Lett 2018; 299:172-181. [PMID: 30312686 DOI: 10.1016/j.toxlet.2018.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 08/31/2018] [Accepted: 10/08/2018] [Indexed: 01/02/2023]
Abstract
The present study investigated the effect of lead (Pb) on bone ultrastructure and chemistry using an in vitro bone model. MC3T3-E1 preosteoblasts were differentiated and treated with lead acetate at 0.4, 2, 10, and 50 μM. No abnormalities in either cell growth or bone nodule formation were observed with the treated dose of lead acetate. However, Pb treatments could significantly increase Pb accumulation in differentiated osteoblast cultures and upregulate expression of Divalent metal transporter 1 (Dmt1) in a dose dependent manner. Pb treatments also altered the expression of osteogenic genes, including secreted phosphoprotein 1, osteocalcin, type I collagen, and osteoprotegerin. Moreover, in mineralized osteoblast cultures, Pb was found to be mainly deposited as Pb salts and oxides, respectively. Ultrastructure analysis revealed Pb localizing with calcium and phosphorus in the mineralized matrix. In mineralizing osteoblast cells, Pb was found in the intracellular calcified vesicles which is one of the bone mineralization mechanisms. Pb was also present in mineral deposits with various shapes and sizes, such as small and large globular or needle-like mineral deposits representing early to mature stages of mineral deposits. Furthermore, Pb was found more in the globular deposits than the needle shaped mineral crystals. Taken together, our observations revealed how Pb incorporates into bone tissue, and showed a close association with bone apatite.
Collapse
Affiliation(s)
- Piyawan Paisrisarn
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Pathumthani 12120, Thailand
| | - Surapun Tepaamorndech
- National Center of Genetic engineering and Biotechnology Center (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Pathumthani 12120, Thailand
| | - Mattaka Khongkow
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Pathumthani 12120, Thailand
| | - Pongtanawat Khemthong
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Pathumthani 12120, Thailand
| | - Panita Kasamechonchung
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Pathumthani 12120, Thailand
| | - Wantana Klysubun
- Synchrotron Light Research Institute, 111 University Avenue, Muang District, Nakhon Ratchasima 30000, Thailand
| | - Tuksadon Wutikhun
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Pathumthani 12120, Thailand
| | - Liping Huang
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA
| | - Kanittha Chantarasakha
- National Center of Genetic engineering and Biotechnology Center (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Pathumthani 12120, Thailand
| | - Suwimon Boonrungsiman
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Pathumthani 12120, Thailand.
| |
Collapse
|
31
|
Zhao W, Shen G, Ren H, Liang D, Yu X, Zhang Z, Huang J, Qiu T, Tang J, Shang Q, Yu P, Wu Z, Jiang X. Therapeutic potential of microRNAs in osteoporosis function by regulating the biology of cells related to bone homeostasis. J Cell Physiol 2018; 233:9191-9208. [PMID: 30078225 DOI: 10.1002/jcp.26939] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are novel regulatory factors that play important roles in numerous cellular processes through the posttranscriptional regulation of gene expression. Recently, deregulation of the miRNA-mediated mechanism has emerged as an important pathological factor in osteoporosis. However, a detailed molecular mechanism between miRNAs and osteoporosis is still not available. In this review, the roles of miRNAs in the regulation of cells related to bone homeostasis as well as miRNAs that deregulate in human or animal are discussed. Moreover, the miRNAs that act as clusters in the biology of cells in the bone microenvironment and the difference of some important miRNAs for bone homeostasis between bone and other organs are mentioned. Overall, miRNAs that contribute to the pathogenesis of osteoporosis and their therapeutic potential are considered.
Collapse
Affiliation(s)
- Wenhua Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gengyang Shen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Ren
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De Liang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhida Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinjing Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Qiu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Tang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peiyuan Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zixian Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaobing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Laboratory Affiliated to National Key Discipline of Orthopaedic and Traumatology of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
32
|
Ferrous and ferric differentially deteriorate proliferation and differentiation of osteoblast-like UMR-106 cells. Biometals 2018; 31:873-889. [DOI: 10.1007/s10534-018-0130-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/10/2018] [Indexed: 12/29/2022]
|