1
|
Luo H, Wang T, Xie Z, Li F, Yang C, Dong W, Wu J, Wang Q, Xu F, Liu J, Zhang F, Peng W. Glucocorticoids regulate the expression of Srsf1 through Hdac4/Foxc1 axis to induce apoptosis of osteoblasts. Commun Biol 2025; 8:566. [PMID: 40186004 PMCID: PMC11971326 DOI: 10.1038/s42003-025-07989-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Further study of the mechanism of glucocorticoid (GC)-induced osteoblast (OB) apoptosis is highly important for the prevention and treatment of GC-induced osteoporosis and osteonecrosis. Serine/arginine-rich splicing factor 1 (Srsf1) expression was downregulated in a dose-dependent manner during GC-induced OB apoptosis. Knockdown of Srsf1 significantly promotes GC-induced OB apoptosis, while overexpression of Srsf1 significantly inhibits GC-induced OB apoptosis. Mechanistically, GC induces the up-regulation of histone deacetylase 4 (Hdac4) in OB, and inhibits the expression of transcription activator forkhead box C1 (Foxc1) by reducing the levels of histone H3 lysine 9 acetylation (H3K9ac) and H3K27ac in the promoter region of Foxc1, thereby down-regulating Srsf1. Next, SRSF1 regulates GC-induced OB apoptosis by regulating Bcl-2 modifying factor (Bmf) alternative splicing. From the perspective of alternative splicing, this study demonstrates that Srsf1 and its regulatory mechanism may serve as a new target for the prevention and treatment of GC-induced osteoporosis and osteonecrosis.
Collapse
Affiliation(s)
- Hong Luo
- Department of Orthopedics and Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Laboratory of Emergency Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Orthopedics, The Affiliated Wudang Hospital of Guizhou Medical University, Guiyang, China
| | - Tao Wang
- Department of Orthopedics and Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Laboratory of Emergency Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhihong Xie
- Department of Orthopedics and Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Laboratory of Emergency Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Fanchao Li
- Laboratory of Emergency Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chengyou Yang
- Laboratory of Emergency Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wentao Dong
- Department of Orthopedics and Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Laboratory of Emergency Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jianhua Wu
- Department of Orthopedics and Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qiang Wang
- Department of Orthopedics and Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Fengyang Xu
- Department of Orthopedics and Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jiong Liu
- Department of Orthopedics, The First People's Hospital of Guiyang, Guiyang, China
| | - Fei Zhang
- Department of Orthopedics and Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.
- Laboratory of Emergency Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Wuxun Peng
- Department of Orthopedics and Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.
- Laboratory of Emergency Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
2
|
Weng Y, Wang X, Tang Y, Du C, Li X, Zhu K, Bao Y, Zeng W, Cai C, Jia B, Yang Z, Tang L. Inhibition of bone morphogenetic protein 4 alleviates angiotensin II-induced abdominal aortic aneurysm by reducing inflammation and endothelial-mesenchymal transition. Atherosclerosis 2025; 403:119134. [PMID: 40081251 DOI: 10.1016/j.atherosclerosis.2025.119134] [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: 02/19/2024] [Revised: 01/21/2025] [Accepted: 02/15/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND AND AIMS Abdominal aortic aneurysm (AAA) is one of the most common fatal macrovascular diseases worldwide which pathogenesis is still not well clarified. In this study, we systematically investigated the alternations of endothelial cell (ECs) functions and phenotypes by single-cell RNA sequencing in angiotensin (Ang) II-induced AAA mice models. METHOD AND RESULTS According to 10 × single-cell sequencing analysis, we revealed that ECs inflammation and endothelial-mesenchymal transition (EndoMT) were involved in the progress of Ang II-induced AAA. Three types of ECs, including Mature ECs (uninjured ECs), EndoMT ECs and Injury & inflammation ECs successively emerged during the progression of AAA. By using pseudotime-trajectory analysis, we speculated bone morphogenetic protein 4 (BMP4) as a candidate gene, participating in Ang II-induced AAA by regulating EndoMT and vascular inflammation. We found that inhibition of BMP4 ameliorated EndoMT and vascular inflammation in Ang II-induced AAA in vivo. In addition, we found that exogenous BMP4 directly promoted the phenotypic transition, inflammation, cell migration and invasion of mouse aortic endothelial cells via PI3K/AKT/mTOR pathways in vitro. Finally, Protein-protein interaction (PPI) analysis and co-immunoprecipitation (Co-IP) revealed that biglycan (BGN) directly combined with BMP4 and promoted the conversion of EndoMT. CONCLUSION Our findings firstly revealed a critical role of BMP4 in AAA progression, which promoted disease progression by inducing EndoMT and reprogramming ECs from anti-inflammatory to proinflammatory phenotype.
Collapse
Affiliation(s)
- Yingzheng Weng
- Department of Medicine, The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310013, China; Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Xihao Wang
- Department of Medicine, The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310013, China
| | - Yimin Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Changqing Du
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Xinyao Li
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Kefu Zhu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Yizhong Bao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Wenping Zeng
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China
| | - Changhong Cai
- Department of Cardiology, Fujian Heart Medical Center, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Bingbing Jia
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China.
| | - Zhouxin Yang
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China.
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, China.
| |
Collapse
|
3
|
Wu J, Ren W, Liu J, Bai X. CUL1 exacerbates glucocorticoid-induced osteoporosis by enhancing ASAP1 ubiquitination. Hormones (Athens) 2025; 24:259-274. [PMID: 39287759 DOI: 10.1007/s42000-024-00599-y] [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: 04/17/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Glucocorticoid-induced osteoporosis is a leading secondary cause of osteoporosis. Cullin-1 (CUL1) levels are abnormally elevated in patients with osteoporosis, but the underlying mechanism remains unclear. The purpose of this study was to elucidate the mechanism of action of CUL1 in a glucocorticoid (dexamethasone, Dex)-induced osteoporosis model. METHODS C57BL/6J mice were intraperitoneally injected with Dex to establish an osteoporosis model. Mouse femur bone injury and bone formation were detected using hematoxylin-eosin or Masson staining. Apoptosis and cell cycle distribution were determined by flow cytometry. Alkaline phosphatase (ALP) activity and calcified nodules were monitored using ALP and Alizarin Red S staining. The molecular mechanism was validated by co-immunoprecipitation (Co-IP) and ubiquitination assays. RESULTS CUL1 expression was enhanced in the Dex-induced osteoporosis mouse model. CUL1 silencing moderated the Dex-induced cell proliferation and osteogenesis inhibition. Moreover, CUL1 promoted the ubiquitination and degradation of ASAP1 via the SKP1-CUL1-F-box (SCF)-FBXW7 complex. CUL1 induced apoptosis and repressed osteogenesis by ASAP1. CUL1 silencing alleviated the Dex-induced osteoporosis in mice. CONCLUSION CUL1 suppressed osteoblast proliferation and osteogenesis by promoting ASAP1 ubiquitination via the SCF-FBXW7 complex in glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- Jun Wu
- Dalian Medical University, No. 9, West Section of Lushun South Road, Dalian, 116041, Liaoning, P.R. China
- Department of Orthopaedics, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Weijian Ren
- Department of Orthopaedics, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Jun Liu
- Department of Orthopaedics, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Xizhuang Bai
- Department of Orthopaedics, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenyang, 110016, Liaoning, People's Republic of China.
| |
Collapse
|
4
|
Wang Z, Liang Q, Lin Z, Li H, Chen X, Zou Z, Mo J. Potential role of formononetin as a novel natural agent in Alzheimer's disease and osteoporosis comorbidity. J Alzheimers Dis 2025; 103:361-371. [PMID: 39828895 DOI: 10.1177/13872877241299104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
BACKGROUND The growing aging population has led to an increase in the prevalence of Alzheimer's disease (AD) and osteoporosis (OP), both of which significantly impair quality of life. The comorbid nature of these conditions suggests a shared genetic etiology, the understanding of which is crucial for developing targeted therapies. OBJECTIVE This study aims to explore the shared genetic etiology underlying AD and OP, using a system biology approach to identify potential therapeutic targets and natural compounds for treatment. METHODS We employed Weighted Gene Co-Expression Network Analysis (WGCNA) with molecular docking strategies to uncover the genetic links between AD and OP. MT2A and CACNA1C were identified as key pleiotropic hub genes potentially linking AD and OP. Molecular docking was utilized to screen for compounds with therapeutic potential, leading to the identification of formononetin as a compound with significant binding affinity to these hub genes. Quantitative real-time PCR (qRT-PCR) validation was conducted to confirm the gene expression changes in disease models. RESULTS Our study indicate that formononetin exhibits strong binding affinity to the identified hub genes, MT2A and CACNA1C. qRT-PCR validation confirmed the upregulation of these genes in disease models, which was mitigated upon treatment with formononetin, suggesting a reversal of disease markers. CONCLUSIONS This study advances our understanding of the genetic intersections between AD and OP and positions formononetin as a promising natural agent for further translational research. Formononetin's multi-target potential makes it a valuable candidate for managing these comorbid conditions, meriting further investigation and development as a therapeutic strategy.
Collapse
Affiliation(s)
- Zhigang Wang
- Clinical Research Center for Neurological Diseases of Guangxi Province, Guilin Medical University, Guilin, China
- Key Laboratory of Brain and Cognition of Guangxi Province, Guilin Medical University, Guilin, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Environmental Exposomics and Entire Lifecycle Health of Guangxi Province, Guilin Medical University, Guilin, China
| | - Qiaoyi Liang
- Clinical Research Center for Neurological Diseases of Guangxi Province, Guilin Medical University, Guilin, China
- Key Laboratory of Brain and Cognition of Guangxi Province, Guilin Medical University, Guilin, China
- Key Laboratory of Environmental Exposomics and Entire Lifecycle Health of Guangxi Province, Guilin Medical University, Guilin, China
| | - Zhaoqiu Lin
- Key Laboratory of Environmental Exposomics and Entire Lifecycle Health of Guangxi Province, Guilin Medical University, Guilin, China
| | - Hongyang Li
- Key Laboratory of Environmental Exposomics and Entire Lifecycle Health of Guangxi Province, Guilin Medical University, Guilin, China
| | - Xin Chen
- Key Laboratory of Environmental Exposomics and Entire Lifecycle Health of Guangxi Province, Guilin Medical University, Guilin, China
| | - Zhenyou Zou
- Biochemistry Department of Purdue University, West Lafayette, IN, USA
| | - Jingxin Mo
- Clinical Research Center for Neurological Diseases of Guangxi Province, Guilin Medical University, Guilin, China
- Key Laboratory of Brain and Cognition of Guangxi Province, Guilin Medical University, Guilin, China
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
5
|
Li Y, Huang Y, Yang H, Li Y, Luo G, Ma Y, Cheng H, Long Y, Xia W, Liu H, Fang H, Du Y, Sun T, Wang K, Zhai X. Composite Scaffold Materials of Nanocerium Oxide Doped with Allograft Bone: Dual Optimization Based on Anti-Inflammatory Properties and Promotion of Osteogenic Mineralization. Adv Healthc Mater 2025; 14:e2403006. [PMID: 39502017 DOI: 10.1002/adhm.202403006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Indexed: 01/15/2025]
Abstract
Spinal fusion technique is widely used in the treatment of lumbar degeneration, cervical instability, disc injury, and spinal deformity. However, it is usually accompanied by a high incidence of fusion failure and pseudoarthrosis, placing higher demands on bone implants. Therefore, materials with good biocompatibility, osteoconductivity, and even induce bone ingrowth, which can be used to improve spinal fusion rate and bone regeneration, have become a hot research topic. Here, ultra-small cerium oxide nanoparticles (CeO2 NPs) are prepared and loaded onto the surface of the homograft bone surface to prepare a composite scaffold AB@PLGA/CeO2. The composite scaffold shows the competitive ability to promote osteoblast differentiation in vitro. In vivo experiments show that AB@PLGA/CeO2 has a good bone enhancement effect. In particular, good biological effects of collagen fiber formation, osteogenic mineralization, and tissue repair are shown in intervertebral implant fusion. Further, transcriptome sequencing confirms that CeO2 NPs promote osteogenic differentiation and mineralization by regulating extracellular matrix (ECM) and collagen formation. Meanwhile, CeO2 NPs can regulate the function of the PI3K-Akt signaling pathway to exert its ability to promote osteogenic differentiation and mineralization and affect p53 and cell cycle signaling pathway to regulate osteogenic differentiation and mineralization. Hence, the proposed scaffold is a promising strategy for intervertebral fusion in the clinic.
Collapse
Affiliation(s)
- Yuqiao Li
- Department of Spine Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Yongkang Huang
- School of Materials Science and Engineering, Tianjin Key Lab for Rare Earth Materials and Applications, Nankai University, Tianjin, 300350, China
| | - Houzhi Yang
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Yuxin Li
- College of Life Sciences, Anhui Normal University, Anhui, 241000, China
| | - Gan Luo
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Yuan Ma
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Haiyang Cheng
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Yubin Long
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
- Department of Spine Surgery, The Central Hospital of Shaoyang, Hunan, 422000, China
| | - Weiwei Xia
- Department of Spine Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Haiying Liu
- Department of Spine Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Huayi Fang
- School of Materials Science and Engineering, Tianjin Key Lab for Rare Earth Materials and Applications, Nankai University, Tianjin, 300350, China
- Shanghat Zhitong Construction Engineering Tethnology Co, LTD
| | - Yaping Du
- School of Materials Science and Engineering, Tianjin Key Lab for Rare Earth Materials and Applications, Nankai University, Tianjin, 300350, China
| | - Tianwei Sun
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Kaifeng Wang
- Department of Spine Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Xinyun Zhai
- School of Materials Science and Engineering, Tianjin Key Lab for Rare Earth Materials and Applications, Nankai University, Tianjin, 300350, China
| |
Collapse
|
6
|
Lu H, Li Z, Zhu L, Xu P, Wang H, Li Y, Zhao W. Fabrication and Temporal Dependency Osteogenic Regulation of Dual-Scale Hierarchical Microstructures on Medical Metal Surface. Adv Healthc Mater 2024; 13:e2402369. [PMID: 39175381 DOI: 10.1002/adhm.202402369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/09/2024] [Indexed: 08/24/2024]
Abstract
The structural characteristics at the interface of bone implants can guide biological regulation. In this study, a dual-scale hierarchical microstructure is proposed and customized using hybrid machining to achieve temporal dependency osteogenic regulation. It is observed that osteoblasts induced by dual-scale hierarchical structure exhibit adequate protrusion development and rapid cell attachment through the modulation of mechanical forces in the cell growth environment, and further promot the upregulation of the cell membrane receptor PDGFR-α, which is related to cell proliferation. Afterward, transcriptomic analysis reveals that during the differentiation stage, the DSH structure regulates cellular signaling cascades primarily through integrin adhesion mechanisms and then accelerates osteogenic differentiation by activating the TGF-β pathway and cAMP signaling pathway. Furthermore, the calcium nodules are preferentially deposited within the lower honeycomb-like channels, thereby endowing the proposed dual-scale hierarchical structure with the potential to induce oriented deposition and improve the long-term stability of the implant.
Collapse
Affiliation(s)
- Hao Lu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Zhijun Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Lida Zhu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Peihua Xu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Hai Wang
- Shenyang Lebuy Vacuum Tech. Co., Ltd, Shenyang, Liaoning, China
| | - Yonghao Li
- Shenyang Lebuy Vacuum Tech. Co., Ltd, Shenyang, Liaoning, China
| | - Weidong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
7
|
Fang Y, Li W, Dong C, Gao B, Guo W, Li M, Jiao Z. Inhibition of SLC40A1 represses osteoblast formation via inducing iron accumulation and activating the PERK/ATF4/CHOP pathway mediated oxidative stress. Redox Rep 2024; 29:2428147. [PMID: 39607819 DOI: 10.1080/13510002.2024.2428147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the effects of solute carrier family 40 member 1 (SLC40A1) on iron accumulation, oxidative stress and differentiation in osteoblasts and potential mechanisms. METHODS Mouse preosteoblastic MC3T3-E1 cells were transfected with the SLC40A1 overexpression vector (oeSLC40A1) and siRNA (siSLC40A1), then cell differentiation was induced via ascorbic acid and β-glycerophosphate. Besides, Ferrostatin-1 (ferroptosis inhibitor) and GSK2606414 (PERK inhibitor) were added with siSLC40A1. RESULTS Fe2+, malondialdehyde (MDA), and reactive oxygen species (ROS) were higher but reduced glutathione (GSH)/oxidized glutathione (GSSG) ratio was lower after siSLC40A1 transfection, while reduced Fe2+ and ROS but elevated GSH/GSSG ratio was observed after oeSLC40A1 transfection. Alkaline phosphatase (ALP) staining, Alizarin Red S (ARS) staining, osteopontin (OPN) and bone morphogenetic protein 2 (BMP2) were lower after siSLC40A1 transfection but were greater after oeSLC40A1 transfection. Furthermore, SLC40A1 negatively regulated the PERK/ATF4/CHOP pathway. Further exploration revealed that Fe2+, MDA, ROS, and the PERK/ATF4/CHOP pathway were attenuated, while GSH/GSSG ratio, ALP staining, ARS staining, and OPN expression were increased after ferrostatin-1 treatment in the siSLC40A1-transfected cells. Similar trends were observed with respect to GSK2606414 treatment with siSLC40A1. CONCLUSION SLC40A1 inhibition suppresses osteoblast formation by facilitating iron accumulation and activating the PERK/ATF4/CHOP pathway-mediated oxidative stress.
Collapse
Affiliation(s)
- Yu Fang
- Department of Orthopedics, Medical Affairs Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Wei Li
- Department of Radiology, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Chongyang Dong
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Binli Gao
- Department of Orthopedics, Medical Affairs Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Wen Guo
- Department of Orthopedics, Medical Affairs Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Mingyu Li
- Department of Orthopedics, Medical Affairs Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Zhichao Jiao
- Department of Orthopedics, Medical Affairs Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People's Republic of China
| |
Collapse
|
8
|
Yizhong B, Chen F, Jin W, Dai J, Mao G, Song B. Sulfated galactofucan from Sargassum fusiforme protects against postmenopausal osteoporosis by regulating bone remodeling. Commun Biol 2024; 7:1471. [PMID: 39516319 PMCID: PMC11549216 DOI: 10.1038/s42003-024-07097-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoporosis is a degenerative bone disease highly prevalent in older women, causing high morbidity and mortality rates. Fourteen kinds of fucoidan were isolated from Sargassum fusiforme through acid (named as SFS), alkaline (SFJ) and water (SFW). SFW was passed through an anion exchange column to obtain SFW-0, SFW-0.5 and SFW-2. SFW-0.5 and SFW-2 were degraded to obtain different sulfate group contents SFW-x-M/S/O (x for 0.5 or 2). We further confirmed SFW-0.5-O was the most effective fraction of SFW. SFW-0.5-O may have alternating backbones of (Gal)n and (Fuc)n, and the main sulfation may be at C2/C3 of the Fuc/Gal residues. SFW-0.5-O inhibition of OC differentiation was associated with IRF-8 signaling; meanwhile, SFW-0.5-O promoted osteoblast differentiation and bone mineral nodule formation. SFW-0.5-O also effectively ameliorated osteoporosis symptom caused by estrogen deprivation in vivo. We uncovered that the fucoidan active fraction SFW-0.5-O demonstrated effective bone protection, may be exploited for osteoporosis therapy.
Collapse
Affiliation(s)
- Bao Yizhong
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, Hangzhou, PR China
| | - Fen Chen
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, PR China
| | - Weihua Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, PR China
| | - Jihua Dai
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, Hangzhou, PR China
| | - Genxiang Mao
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, Hangzhou, PR China.
| | - Boshan Song
- Department of Orthopaedic Surgery, Zhejiang Hospital, Hangzhou, PR China.
| |
Collapse
|
9
|
Hu J, Song Y, Zhang Y, Yang P, Chen S, Wu Z, Zhang J. Catalpol Enhances Osteogenic Differentiation of Human Periodontal Stem Cells and Modulates Periodontal Tissue Remodeling in an Orthodontic Tooth Movement Rat Model. Drug Des Devel Ther 2024; 18:4943-4960. [PMID: 39525045 PMCID: PMC11546164 DOI: 10.2147/dddt.s482969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose This study examines the effects and mechanisms of catalpol (CAT) on the proliferation and osteogenic differentiation of cultured human periodontal ligament stem cells (hPDLSCs) in vitro and assesses the impact of CAT on periodontal remodeling in vivo using an orthodontic tooth movement (OTM) model in rats. Methods hPDLSCs were cultured in a laboratory setting, and their proliferation and osteogenic differentiation were assessed using the Cell-counting Kit-8 (CCK-8), Alizarin Red Staining (ARS), quantitative calcium assay, alkaline phosphatase (ALP) staining and activity assay, and immunofluorescence assay. Additionally, the expression of collagen type 1 (COL-1), ALP, and runt-related transcription factor-2 (RUNX-2) was evaluated through qRT-PCR and Western blot analysis. To verify the function of the estrogen receptor-α (ER-α)-mediated phosphatidylinositol-3-kinase-protein kinase B (PI3K/AKT) pathway in this mechanism, LY294002 (a PI3K signaling pathway inhibitor) and the ER-α specific inhibitor methyl-piperidine-pyrazole (MPP) were used. The osteogenic markers ER-α, AKT, and p-AKT (phosphoprotein kinase B) were identified through Western blot analysis. Eighteen male Sprague-Dawley rats were assigned to two groups randomly: a CAT group receiving CAT and a control group receiving an equivalent volume of saline. Micro-computed tomography (micro-CT) analysis was employed to evaluate tooth movement and changes in alveolar bone structure. Morphological changes in the periodontal tissues between the roots were investigated using hematoxylin and eosin (HE) staining and tartaric-resistant acid phosphatase (TRAP) staining. The expression of COL-1, RUNX-2, and nuclear factor-κB (NF-κB) ligand (RANKL) was assessed through immunohistochemical staining (IHC) to evaluate periodontal tissue remodeling. Tests were analyzed using GraphPad Prism 8 software. Differences among more than two groups were analyzed by one-way or two-way analysis of variance (ANOVA) followed by the Tukey's test. Values of p < 0.05 were regarded as statistically significant. Results In vitro experiments demonstrated that 10 μM CAT significantly promoted the proliferation, ALP activity, and calcium nodule formation of hPDLSCs, with a notable increase in the expression of COL-1, ALP, RUNX-2, ER-α, and p-AKT. The PI3K/AKT pathway was inhibited by LY294002, and further analysis using MPP suggested that ER-α mediated this effect. In vivo, experiments indicated that CAT enhanced the expression of COL-1 and RUNX-2 on the tension side of rat tooth roots, reduced the number of osteoclasts on the compression side, inhibited RANKL expression, and suppressed OTM. Conclusion CAT can promote hPDLSCs proliferation and osteogenic differentiation in vitro through the ER-α/PI3K/AKT pathway and enhance periodontal tissue remodeling in vivo using OTM models. These findings suggest the potential for the clinical application of catalpol in preventing relapse following OTM.
Collapse
Affiliation(s)
- Jing Hu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Yang Song
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Yuxing Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Peng Yang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Siyu Chen
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Zhaoyan Wu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Jun Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
10
|
Jin R, Li C, Yang Y, Xie J. AEBP1 restores osteoblastic differentiation under dexamethasone treatment by activating PI3K/AKT signalling. Clin Exp Pharmacol Physiol 2024; 51:e13923. [PMID: 39358837 DOI: 10.1111/1440-1681.13923] [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: 04/28/2024] [Revised: 07/23/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024]
Abstract
Adipocyte enhancer-binding protein 1 (AEBP1) is closely implicated in osteoblastic differentiation and bone fracture; this research aimed to investigate the effect of AEBP1 on restoring osteoblastic differentiation under dexamethasone (Dex) treatment, and its interaction with the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway. Pre-osteoblastic MC3T3-E1 cells were cultured in osteogenic medium and treated by Dex to mimic steroid-induced osteonecrosis cellular model. They were then further transfected with control or AEBP1-overexpressed lentiviral vectors. Finally, cells were treated with the PI3K inhibitor LY294002, with or without AEBP1-overexpressed lentiviral vectors. AEBP1 expression showed a downward trend in MC3T3-E1 cells under Dex treatment in a dose-dependent manner. AEBP1-overexpressed lentiviral vectors increased relative cell viability, alkaline phosphatase (ALP) staining, Alizarin red staining and osteoblastic differentiation markers including osteocalcin (OCN), osteopontin (OPN), collagen type I alpha 1 (COL1A1), runt-related transcription factor 2 (RUNX2) and bone morphogenetic protein 2 (BMP2), but decreased cell apoptosis rate in MC3T3-E1 cells under Dex treatment; besides, AEBP1-overexpressed lentiviral vectors positively regulated p-PI3K and p-AKT expressions. Furthermore, LY294002 treatment decreased relative cell viability, Alizarin red staining, osteoblastic differentiation markers including OCN, OPN, RUNX2 and BMP, increased cell apoptosis rate and did not affect ALP staining in MC3T3-E1 cells under Dex treatment; meanwhile, LY294002 treatment weakened the effect of AEBP1 overexpression vectors on the above cell functions. AEBP1 restores osteoblastic differentiation under Dex treatment by activating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Rilong Jin
- Center for Sport Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Li
- Center for Sport Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yute Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Xie
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Qu Z, Zhao S, Zhang Y, Wang X, Yan L. Natural Compounds for Bone Remodeling: Targeting osteoblasts and relevant signaling pathways. Biomed Pharmacother 2024; 180:117490. [PMID: 39332184 DOI: 10.1016/j.biopha.2024.117490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024] Open
Abstract
In the process of bone metabolism and bone remodeling, bone marrow mesenchymal stem cells (BM-MSCs) differentiate into osteoblasts (OBs) under certain conditions to enable the formation of new bone, and normal bone reconstruction and pathological bone alteration are closely related to the differentiation and proliferation functions of OBs. Osteogenic differentiation of BM-MSCs involves multiple signaling pathways, which function individually but interconnect intricately to form a complex signaling regulatory network. Natural compounds have fewer adverse effects than chemically synthesized drugs, optimize bone health, and are more suitable for long-term use. In this paper, we focus on OBs, summarize the current research progress of signaling pathways related to OBs differentiation, and review the molecular mechanisms by which chemically synthesized drugs with potential anti-osteoporosis properties regulate OBs-mediated bone formation.
Collapse
Affiliation(s)
- Zechao Qu
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Songchuan Zhao
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yong Zhang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaohao Wang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Yan
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
12
|
Jia F, Guan J, Wang J, Li M, Zhang Y, Xie L, Han P, Lin H, Huang X, Lan J, Huang Y. Zinc and melatonin mediated antimicrobial, anti-inflammatory, and antioxidant coatings accelerate bone defect repair. Colloids Surf B Biointerfaces 2024; 245:114335. [PMID: 39461184 DOI: 10.1016/j.colsurfb.2024.114335] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Inflammation and bacterial infection are important causes of implant failure, and the development of multifunctional titanium surfaces to address these issues is an effective means of treating infected bone defects. In this study, polyphenols (EGCG) and Zn2+ were first loaded onto the titanium surface to construct an EGCG/Zn2+ polyphenol metal network coating. Then melatonin (MT) was loaded into the EGCG/Zn2+ network structure to prepare the EGCG/Zn2+/MT composite coating. The results proved that the EGCG/Zn2+/MT coating had good mechanical properties, hydrophilicity, corrosion resistance and bioactivity. In vitro, the inhibition rates of EGCG/Zn2+/MT against E. coli and S. aureus were about 97 % and 81 %, respectively. In vitro experiments revealed that EGCG/Zn2+/MT could regulate the polarization of macrophages (RAW264.7) to M2 type, could induce vascularization of human umbilical vein endothelial cells (HUVEC), and could promote the differentiation of pro-osteoblasts (MC3T3-E1) to osteogenesis. Meanwhile, EGCG/Zn2+/MT achieved effective ROS scavenging within HUVEC and MC3T3-E1. In vivo experiments demonstrated that the EGCG/Zn2+/MT coatings possessed favorable biosafety, anti-inflammatory, antimicrobial, and bone repair capabilities. This study provides a simple and versatile strategy for designing multifunctional surfaces with both antimicrobial, anti-inflammatory, antioxidant, angiogenic and osteogenic properties.
Collapse
Affiliation(s)
- Fengzhen Jia
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Jiaxin Guan
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Jiali Wang
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Meiyu Li
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Yasi Zhang
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China
| | - Lei Xie
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Pengde Han
- School of Materials Science and Engineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - He Lin
- School of Chemistry and Materials Science, Ludong University, Yantai, 264025, China
| | - Xiao Huang
- Hunan Provincial Key Laboratory of Dong Medicine, Ethnic Medicine Research Center, Hunan University of Medicine, Huaihua 418000, PR China.
| | - Jinping Lan
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China.
| | - Yong Huang
- College of Lab Medicine, Life Science Research Centre, Hebei North University, Zhangjiakou 075000, China.
| |
Collapse
|
13
|
Yang N, Li M, Li X, Wu L, Wang W, Xu Y, Wang Z, Zhu C, Geng D. MAGL blockade alleviates steroid-induced femoral head osteonecrosis by reprogramming BMSC fate in rat. Cell Mol Life Sci 2024; 81:418. [PMID: 39368012 PMCID: PMC11455816 DOI: 10.1007/s00018-024-05443-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/01/2024] [Accepted: 09/08/2024] [Indexed: 10/07/2024]
Abstract
The leading cause of steroid-induced femoral head osteonecrosis (ONFH) is the imbalance of bone homeostasis. Bone marrow-derived mesenchymal stem cell (BMSC) differentiation and fate are closely associated with bone homeostasis imbalance. Blocking monoacylglycerol lipase (MAGL) could effectively ameliorate ONFH by mitigating oxidative stress and apoptosis in BMSCs induced by glucocorticoids (GC). Nevertheless, whether MAGL inhibition can modulate the balance during BMSC differentiation, and therefore improve ONFH, remains elusive. Our study indicates that MAGL inhibition can effectively rescue the enhanced BMSC adipogenic differentiation caused by GC and promote their differentiation toward osteogenic lineages. Cannabinoid receptor 2 (CB2) is the direct downstream target of MAGL in BMSCs, rather than cannabinoid receptor 1(CB1). Using RNA sequencing analyses and a series of in vitro experiments, we confirm that the MAGL blockade-induced enhancement of BMSC osteogenic differentiation is primarily mediated by the phosphoinositide 3-kinases (PI3K)/ the serine/threonine kinase (AKT)/ (glycogen synthase kinase-3 beta) GSK3β pathway. Additionally, MAGL blockade can also reduce GC-induced bone resorption by directly suppressing osteoclastogenesis and indirectly reducing the expression of receptor activator of nuclear factor kappa-Β ligand (RANKL) in BMSCs. Thus, our study proposes that the therapeutic effect of MAGL blockade on ONFH is partly mediated by restoring the balance of bone homeostasis and MAGL may be an effective therapeutic target for ONFH.
Collapse
Affiliation(s)
- Ning Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215006, China
- Department of Orthopaedics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Meng Li
- Department of Orthopaedics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Xuefeng Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Lunan Wu
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, Hefei, 230001, China
| | - Wenzhi Wang
- Department of Orthopaedics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yaozeng Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Zhen Wang
- Department of Orthopaedics, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215000, China
| | - Chen Zhu
- Department of Orthopaedics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215006, China.
| |
Collapse
|
14
|
Zhao C, Zhu S, Liang Y, Xu S. Asymmetric Osteopenia in Adolescent Idiopathic Scoliosis Based on Hounsfield Unit of Computed Tomography. Int J Gen Med 2024; 17:3945-3953. [PMID: 39263592 PMCID: PMC11389713 DOI: 10.2147/ijgm.s478933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
Purpose To characterize the asymmetrical loss of bone mass and identify the association between scoliosis and osteopenia in patients with adolescent idiopathic scoliosis (AIS). Methods Demographic information, Cobb angle, and Hounsfield unit (HU) of the neutral vertebra (NV) and apical vertebra (apex) of the major curve were collected retrospectively in 54 AIS patients. For 84 control subjects, HU values were measured at T12 and L5. Propensity score matching was performed to balance the interference of age and BMI. Results In the AIS group, the concave and convex lateral HU of the NV and the convex lateral HU of the apex were negatively correlated with the Cobb angle. The AIS patients had lower bilateral HU. The mean HU and the apex-convex HU were also lower in the AIS group, while the apex-concave HU was slightly higher. After matching, the apex-convex HU of the AIS group remained lower, while the apex-concave HU was higher. Conclusion Patients with AIS exhibit osteopenia, particularly on the convex side. The severity of scoliosis was found to be directly proportional to the severity of bone loss and the degree of bilateral osteopenia asymmetry. Appropriate intervention for bone loss may be able to curb the progression of scoliosis.
Collapse
Affiliation(s)
- Chong Zhao
- Department of Spinal Surgery, Peking University People's Hospital, Peking University, Beijing, People's Republic of China
| | - Shuaiqi Zhu
- Department of Spinal Surgery, Peking University People's Hospital, Peking University, Beijing, People's Republic of China
| | - Yan Liang
- Department of Spinal Surgery, Peking University People's Hospital, Peking University, Beijing, People's Republic of China
| | - Shuai Xu
- Department of Spinal Surgery, Peking University People's Hospital, Peking University, Beijing, People's Republic of China
| |
Collapse
|
15
|
Sun Q, Xu L, Hu Z, Liu J, Yu T, Li M, Zhang S, Shi F. Melatonin Regulates Osteoblast Differentiation through the m6A Reader hnRNPA2B1 under Simulated Microgravity. Curr Issues Mol Biol 2024; 46:9624-9638. [PMID: 39329924 PMCID: PMC11430354 DOI: 10.3390/cimb46090572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Recent studies have confirmed that melatonin and N6-methyladenosine (m6A) modification can influence bone cell differentiation and bone formation. Melatonin can also regulate a variety of biological processes through m6A modification. Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2B1) serves as a reader of m6A modification. In this study, we used the hindlimb unloading model as an animal model of bone loss induced by simulated microgravity and used 2D clinorotation to simulate a microgravity environment for cells on the ground. We found that hnRNPA2B1 was downregulated both in vitro and in vivo during simulated microgravity. Further investigations showed that hnRNPA2B1 could promote osteoblast differentiation and that overexpression of hnRNPA2B1 attenuated the suppression of osteoblast differentiation induced by simulated microgravity. We also discovered that melatonin could promote the expression of hnRNPA2B1 under simulated microgravity. Moreover, we found that promotion of osteoblast differentiation by melatonin was partially dependent on hnRNPA2B1. Therefore, this research revealed, for the first time, the role of the melatonin/hnRNPA2B1 axis in osteoblast differentiation under simulated microgravity. Targeting this axis may be a potential protective strategy against microgravity-induced bone loss and osteoporosis.
Collapse
Affiliation(s)
- Quan Sun
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Liqun Xu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Zebing Hu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Jingchun Liu
- No. 5 Cadet Regiment, School of Basic Medical Sciences, Air Force Medical University, Xi’an 710032, China; (J.L.); (T.Y.)
| | - Tingfei Yu
- No. 5 Cadet Regiment, School of Basic Medical Sciences, Air Force Medical University, Xi’an 710032, China; (J.L.); (T.Y.)
| | - Meng Li
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Fei Shi
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| |
Collapse
|
16
|
Wang W, Gong Z, Wang K, Tian M, Zhang Y, Li X, You X, Wu J. Activation of the BMP2-SMAD1-CGRP pathway in dorsal root ganglia contributes to bone cancer pain in a rat model. Heliyon 2024; 10:e27350. [PMID: 38496903 PMCID: PMC10944225 DOI: 10.1016/j.heliyon.2024.e27350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024] Open
Abstract
Peripheral nerve remodeling and sensitization are involved in cancer-related bone pain. As a member of the transforming growth factor-β class, bone morphogenetic protein 2 (BMP2) is recognized to have a role in the development of the neurological and skeletal systems. Our previous work showed that BMP2 is critical for bone cancer pain (BCP) sensitization. However, the mechanism remains unknown. In the current study, we demonstrated a substantial increase in BMP2 expression in the dorsal root ganglia (DRG) in a rat model of BCP. Knockdown of BMP2 expression ameliorated BCP in rats. Furthermore, the DRG neurons of rats with BCP expressed higher levels of calcitonin gene-related peptide (CGRP), and BCP was successfully suppressed by intrathecal injection of a CGRP receptor blocker (CGRP8-37). Downregulation of BMP2 expression reduced the expression of CGRP in the DRG of rats with BCP and relieved pain behavior. Moreover, we revealed that upregulation of CGRP expression in the DRG may be induced by activation of the BMPR/Smad1 signaling pathway. These findings suggest that BMP2 contributes to BCP by upregulating CGRP in DRG neurons via activating BMPR/Smad1 signaling pathway and that therapeutic targeting of the BMP2-Smad1-CGRP pathway may ameliorate BCP in the context of advanced cancer.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200030, China
| | - Zhihao Gong
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200030, China
| | - Kai Wang
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200030, China
| | - Mi Tian
- Department of Intensive Care Medicine, HuaShan Hospital, Fudan University, Shanghai 200040, China
| | - Yuxin Zhang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200030, China
| | - Xin Li
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Xingji You
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Jingxiang Wu
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200030, China
| |
Collapse
|
17
|
Lin C, Jiang H, Lou C, Wang W, Cai T, Lin Z, Jiang L, Lin S, Xue X, Pan X. Asiatic acid prevents glucocorticoid-induced femoral head osteonecrosis via PI3K/AKT pathway. Int Immunopharmacol 2024; 130:111758. [PMID: 38422771 DOI: 10.1016/j.intimp.2024.111758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/22/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Glucocorticoid-induced osteonecrosis of the femoral head (GIONFH) represents a predominant etiology of non-traumatic osteonecrosis, imposing substantial pain, restricting hip mobility, and diminishing overall quality of life for affected individuals. Centella asiatica (L.) Urb. (CA), an herbal remedy deeply rooted in traditional oriental medicine, has exhibited noteworthy therapeutic efficacy in addressing inflammation and facilitating wound healing. Drawing from CA's historical applications, its anti-inflammatory, anti-apoptotic, and antioxidant attributes may hold promise for managing GIONFH. Asiatic acid (AA), a primary constituent of CA, has been substantiated as a key contributor to its anti-apoptotic, antioxidant, and anti-inflammatory capabilities, showcasing a close association with orthopedic conditions. For the investigation of whether AA could alleviate GIONFH through suppressing oxidative stress, apoptosis, and to delve into its potential cellular and molecular mechanisms, the connection between AA and disease was analyzed through network pharmacology. DEX-induced apoptosis in rat osteoblasts and GIONFH in rat models, got utilized for the verification in vitro/vivo, on underlying mechanism of AA in GIONFH. Network pharmacology analysis reveals a robust correlation between AA and GIONFH in multiple target genes. AA has demonstrated the inhibition of DEX-induced osteoblast apoptosis by modulating apoptotic factors like BAX, BCL-2, Cleaved-caspase3, and cleaved-caspase9. Furthermore, it effectively diminishes the ROS overexpression and regulates oxidative stress through mitochondrial pathway. Mechanistic insights suggest that AA's therapeutic effects involve phosphatidylinositol 3-kinase/Protein kinase B (PI3K/AKT) pathway activation. Additionally, AA has exhibited its potential to ameliorate GIONFH progression in rat models. Our findings revealed that AA mitigated DEX-induced osteoblast apoptosis and oxidative stress through triggering PI3K/AKT pathway. Also, AA can effectively thwart GIONFH occurrence and development in rats.
Collapse
Affiliation(s)
- Chihao Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hongyi Jiang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chao Lou
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Weidan Wang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tingwen Cai
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhongnan Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Liting Jiang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shida Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xinghe Xue
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xiaoyun Pan
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
18
|
Du X, Zang C, Wang Q. Cyclin A1 (CCNA1) inhibits osteoporosis by suppressing transforming growth factor-beta (TGF-beta) pathway in osteoblasts. BMC Musculoskelet Disord 2024; 25:206. [PMID: 38454404 PMCID: PMC10919014 DOI: 10.1186/s12891-024-07303-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Osteoporosis is a genetic disease caused by the imbalance between osteoblast-led bone formation and osteoclast-induced bone resorption. However, further gene-related pathogenesis remains to be elucidated. METHODS The aberrant expressed genes in osteoporosis was identified by analyzing the microarray profile GSE100609. Serum samples of patients with osteoporosis and normal group were collected, and the mRNA expression of candidate genes was detected by quantitative real-time polymerase chain reaction (qRT-PCR). The mouse cranial osteoblast MC3T3-E1 cells were treated with dexamethasone (DEX) to mimic osteoporosis in vitro. Alizarin Red staining and alkaline phosphatase (ALP) staining methods were combined to measure matrix mineralization deposition of MC3T3-E1 cells. Meanwhile, the expression of osteogenesis related genes including alkaline phosphatase (ALP), osteocalcin (OCN), osteopontin (OPN), Osterix, and bone morphogenetic protein 2 (BMP2) were evaluated by qRT-PCR and western blotting methods. Then the effects of candidate genes on regulating impede bone loss caused by ovariectomy (OVX) in mice were studied. RESULTS Cyclin A1 (CCNA1) was found to be significantly upregulated in serum of osteoporosis patients and the osteoporosis model cells, which was in line with the bioinformatic analysis. The osteogenic differentiation ability of MC3T3-E1 cells was inhibited by DEX treatment, which was manifested by decreased Alizarin Red staining intensity, ALP staining intensity, and expression levels of ALP, OCN, OPN, Osterix, and BMP2. The effects of CCNA1 inhibition on regulating osteogenesis were opposite to that of DEX. Then, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis demonstrated that genes negatively associated with CCNA1 were enriched in the TGF-beta signaling pathway. Inhibitor of TGF-beta signaling pathway partly reversed osteogenesis induced by suppressed CCNA1. Furthermore, suppressed CCNA1 relieved bone mass of OVX mice in vivo. CONCLUSION Downregulation of CCNA1 could activate TGF-beta signaling pathway and promote bone formation, thus playing a role in treatment of osteoporosis.
Collapse
Affiliation(s)
- Xiao Du
- Department of Orthopedics, Beijing Geriatric Hospital, No.118 Hot Spring Road, Haidian District 100095, Beijing, China
| | - Chuanyi Zang
- Department of Orthopedics, Beijing Geriatric Hospital, No.118 Hot Spring Road, Haidian District 100095, Beijing, China
| | - Qinglei Wang
- Department of Orthopedics, Beijing Geriatric Hospital, No.118 Hot Spring Road, Haidian District 100095, Beijing, China.
| |
Collapse
|
19
|
Cai R, Shan Y, Du F, Miao Z, Zhu L, Hang L, Xiao L, Wang Z. Injectable hydrogels as promising in situ therapeutic platform for cartilage tissue engineering. Int J Biol Macromol 2024; 261:129537. [PMID: 38278383 DOI: 10.1016/j.ijbiomac.2024.129537] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/01/2024] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
Injectable hydrogels are gaining prominence as a biocompatible, minimally invasive, and adaptable platform for cartilage tissue engineering. Commencing with their synthesis, this review accentuates the tailored matrix formulations and cross-linking techniques essential for fostering three-dimensional cell culture and melding with complex tissue structures. Subsequently, it spotlights the hydrogels' enhanced properties, highlighting their augmented functionalities and broadened scope in cartilage tissue repair applications. Furthermore, future perspectives are advocated, urging continuous innovation and exploration to surmount existing challenges and harness the full clinical potential of hydrogels in regenerative medicine. Such advancements are crucial for validating the long-term efficacy and safety of hydrogels, positioning them as a promising direction in regenerative medicine to address cartilage-related ailments.
Collapse
Affiliation(s)
- Rong Cai
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Yisi Shan
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Fengyi Du
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212013, China
| | - Zhiwei Miao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Like Zhu
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Li Hang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Long Xiao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| | - Zhirong Wang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| |
Collapse
|
20
|
Liu LL, Liu ZR, Cao LJ, Wang J, Huang SM, Hu SG, Yang YZ, Li DS, Cao WW, Zeng QB, Huang S, Wu Q, Xiao JH, Liu WY, Xiao YS. Iron accumulation induced by hepcidin1 knockout accelerates the progression of aging osteoporosis. J Orthop Surg Res 2024; 19:59. [PMID: 38216929 PMCID: PMC10785403 DOI: 10.1186/s13018-024-04535-z] [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: 10/20/2023] [Accepted: 01/04/2024] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVE Iron accumulation is associated with osteoporosis. This study aims to explore the effect of chronic iron accumulation induced by hepcidin1 deficiency on aging osteoporosis. METHODS Iron accumulation in hepcidin1 knockout aging mice was assessed by atomic absorption spectroscopy and Perl's staining. Bone microarchitecture was observed using Micro-CT. Hepcidin, ferritin, oxidative stress, and markers of bone turnover in serum were detected by enzyme-linked immunosorbent assay. Bone formation and resorption markers were measured by real-time quantitative PCR. Cell aging was induced by D-galactose treatment. CCK-8, flow cytometry, EdU assays, and Alizarin red staining were performed to reveal the role of hepcidin1 knockout in cell model. Iron Colorimetric Assay Kit and western blot were applied to detect iron and ferritin levels in cells, respectively. RESULTS In hepcidin1-knockout mice, the ferritin and iron contents in liver and tibia were significantly increased. Iron accumulation induced by hepcidin1 knockout caused a phenotype of low bone mass and deteriorated bone microarchitecture. Osteogenic marker was decreased and osteoclast marker was increased in mice, accompanied by increased oxidative stress level. The mRNA expression levels of osteoclast differentiation markers (RANKL, Mmp9, OPG, Trap, and CTSK) were up-regulated, while bone formation markers (OCN, ALP, Runx2, SP7, and Col-1) were down-regulated in model group, compared to wild type mice. In vitro, hepcidin1 knockdown inhibited proliferation and osteogenic differentiation, while promoted apoptosis, with increased levels of iron and ferritin. CONCLUSION Iron accumulation induced by hepcidin1 deficiency aggravates the progression of aging osteoporosis via inhibiting osteogenesis and promoting osteoclast genesis.
Collapse
Affiliation(s)
- Lu-Lin Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China
| | - Zhong-Rui Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China
| | - Lu-Jun Cao
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Jun Wang
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - San-Ming Huang
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Shui-Gen Hu
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Yi-Zhong Yang
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Dong-Sheng Li
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Wei-Wei Cao
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Qing-Bao Zeng
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Sheng Huang
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Qiong Wu
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Jian-Hua Xiao
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China
| | - Wu-Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China
| | - Yao-Sheng Xiao
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China.
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
21
|
Bagherifard A, Hosseinzadeh A, Koosha F, Sheibani M, Karimi-Behnagh A, Reiter RJ, Mehrzadi S. Melatonin and bone-related diseases: an updated mechanistic overview of current evidence and future prospects. Osteoporos Int 2023; 34:1677-1701. [PMID: 37393580 DOI: 10.1007/s00198-023-06836-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/16/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE Bone diseases account for an enormous cost burden on health systems. Bone disorders are considered as age-dependent diseases. The aging of world population has encouraged scientists to further explore the most effective preventive modalities and therapeutic strategies to overcome and reduce the high cost of bone disorders. Herein, we review the current evidence of melatonin's therapeutic effects on bone-related diseases. METHODS This review summarized evidences from in vitro, in vivo, and clinical studies regarding the effects of melatonin on bone-related diseases, with a focus on the molecular mechanisms. Electronically, Scopus and MEDLINE®/PubMed databases were searched for articles published on melatonin and bone-related diseases from inception to June 2023. RESULTS The findings demonstrated that melatonin has beneficial effect in bone- and cartilage-related disorders such as osteoporosis, bone fracture healing, osteoarthritis, and rheumatoid arthritis, in addition to the control of sleep and circadian rhythms. CONCLUSION A number of animal and clinical studies have indicated that various biological effects of melatonin may suggest this molecule as an effective therapeutic agent for controlling, diminishing, or suppressing bone-related disorders. Therefore, further clinical studies are required to clarify whether melatonin can be effective in patients with bone-related diseases.
Collapse
Affiliation(s)
- Abolfazl Bagherifard
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, 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
| | - Mohammad Sheibani
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Russel J Reiter
- Department of Cellular and Structural Biology, Long School of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Li Q, Tian C, Liu X, Li D, Liu H. Anti-inflammatory and antioxidant traditional Chinese Medicine in treatment and prevention of osteoporosis. Front Pharmacol 2023; 14:1203767. [PMID: 37441527 PMCID: PMC10335577 DOI: 10.3389/fphar.2023.1203767] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
A metabolic bone disorder called osteoporosis is characterized by decreased bone mass and compromised microarchitecture. This condition can deteriorate bones and raise the risk of fractures. The two main causes of osteoporosis are an increase in osteoclast activity or quantity and a decrease in osteoblast viability. Numerous mechanisms, including estrogen shortage, aging, chemical agents, and decreased mechanical loads, have been linked to osteoporosis. Inflammation and oxidative stress have recently been linked to osteoporosis, according to an increasing number of studies. The two primary medications used to treat osteoporosis at the moment are bisphosphonates and selective estrogen receptor modulators (SERMs). These medications work well for osteoporosis brought on by aging and estrogen deprivation, however, they do not target inflammation and oxidative stress-induced osteoporosis. In addition, these drugs have some limitations that are attributed to various side effects that have not been overcome. Traditional Chinese medicine (TCM) has been applied in osteoporosis for many years and has a high safety profile. Therefore, in this review, literature related to botanical drugs that have an effect on inflammation and oxidative stress-induced osteoporosis was searched for. Moreover, the pharmacologically active ingredients of these herbs and the pathways were discussed and may contribute to the discovery of more safe and effective drugs for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Qian Li
- Laboratory of Metabolic Abnormalities and Vascular Aging, Liyuan Hospital Affiliated to Huazhong University of Science and Technology, Department of Integrated Chinese and Western Medicine, City Wuhan, Hubei Province, China
| | - Ciqiu Tian
- Hubei University of Chinese Medicine, City Wuhan, Hubei Province, China
| | - Xiangjie Liu
- Liyuan Hospital Affiliated to Huazhong University of Science and Technology, Geriatric Department, City Wuhan, Hubei Province, China
| | - Dinglin Li
- Laboratory of Metabolic Abnormalities and Vascular Aging, Liyuan Hospital Affiliated to Huazhong University of Science and Technology, Department of Integrated Chinese and Western Medicine, City Wuhan, Hubei Province, China
| | - Hao Liu
- Laboratory of Metabolic Abnormalities and Vascular Aging, Liyuan Hospital Affiliated to Huazhong University of Science and Technology, Department of Integrated Chinese and Western Medicine, City Wuhan, Hubei Province, China
| |
Collapse
|
23
|
Melatonin Repairs Osteoporotic Bone Defects in Iron-Overloaded Rats through PI3K/AKT/GSK-3 β/P70S6k Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7718155. [PMID: 36703914 PMCID: PMC9873465 DOI: 10.1155/2023/7718155] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 10/05/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023]
Abstract
It was found recently that iron overload can cause osteoporosis in rats. Through in vitro and in vivo experimentations, the purpose of the present study was to validate and confirm the inhibitory effects of melatonin on iron death of osteoporosis and its role in bone microstructure improvements. Melatonin (100 mol/L) was administered to MC3T3-E1 cells induced by iron overload in vitro for 48 hours. The expression of cleaved caspase-3 and cleaved PARP and the production of ROS (reactive oxygen species) and mitochondrial damage were all exacerbated by iron overload. On the other hand, melatonin restored these impacts in MC3T3-E1 cells produced by iron overload. By evaluating the expression of PI3K/AKT/GSK-3β/P70S6k signaling pathway-related proteins (RUNX2, BMP2, ALP, and OCN) using RT-PCR and Western blot, osteogenic-related proteins were identified. Alizarin red S and alkaline phosphatase were utilized to evaluate the osteogenic potential of MC3T3-E1 cells. Melatonin significantly improved the osteogenic ability and phosphorylation rates of PI3K, AKT, GSK-3β, and P70S6k in iron overload-induced MC3T3-E1 cells. In vivo, melatonin treated iron overload-induced osteoporotic bone defect in rats. Rat skeletal microstructure was observed using micro-CT and bone tissue pathological section staining. ELISA was utilized to identify OCN, PINP, CTX-I, and SI in the serum of rats. We discovered that melatonin increased bone trabecular regeneration and repair in osteoporotic bone defects caused by iron overload. In conclusion, melatonin enhanced the osteogenic ability of iron overload-induced MC3T3-E1 cells by activating the PI3K/AKT/GSK-3β/P70S6k signaling pathway and promoting the healing of iron overload-induced osteoporotic bone defects in rats.
Collapse
|
24
|
Yang K, Qiu X, Cao L, Qiu S. The role of melatonin in the development of postmenopausal osteoporosis. Front Pharmacol 2022; 13:975181. [PMID: 36278157 PMCID: PMC9585202 DOI: 10.3389/fphar.2022.975181] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/26/2022] [Indexed: 11/28/2022] Open
Abstract
Melatonin is an important endogenous hormone that modulates homeostasis in the microenvironment. Recent studies have indicated that serum melatonin levels are closely associated with the occurrence and development of osteoporosis in postmenopausal women. Exogenous melatonin could also improve bone mass and increase skeletal strength. To determine the underlying mechanisms of melatonin in the prevention and treatment of postmenopausal osteoporosis, we performed this review to analyze the role of melatonin in bone metabolism according to its physiological functions. Serum melatonin is related to bone mass, the measurement of which is a potential method for the diagnosis of osteoporosis. Melatonin has a direct effect on bone remodeling by promoting osteogenesis and suppressing osteoclastogenesis. Melatonin also regulates the biological rhythm of bone tissue, which benefits its osteogenic effect. Additionally, melatonin participates in the modulation of the bone microenvironment. Melatonin attenuates the damage induced by oxidative stress and inflammation on osteoblasts and prevents osteolysis from reactive oxygen species and inflammatory factors. As an alternative drug for osteoporosis, melatonin can improve the gut ecology, remodel microbiota composition, regulate substance absorption and maintain metabolic balance, all of which are beneficial to the health of bone structure. In conclusion, our review systematically demonstrates the effects of melatonin on bone metabolism. Based on the evidence in this review, melatonin will play a more important role in the diagnosis, prevention and treatment of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Keda Yang
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
| | - Xueshan Qiu
- Department of Pathology, The First Affiliated Hospital of China Medical University and College of Basic Medical Sciences Shenyang, Shenyang, Liaoning, China
| | - Lili Cao
- Department of Medical Oncology, First Hospital of China Medical University, Shenyang, China
- *Correspondence: Lili Cao, ; Shui Qiu,
| | - Shui Qiu
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
- *Correspondence: Lili Cao, ; Shui Qiu,
| |
Collapse
|
25
|
Zheng F, Zhang F, Wang F. Inhibition of miR‑98‑5p promotes high glucose‑induced suppression of preosteoblast proliferation and differentiation via the activation of the PI3K/AKT/GSK3β signaling pathway by targeting BMP2. Mol Med Rep 2022; 26:292. [PMID: 35904181 PMCID: PMC9366150 DOI: 10.3892/mmr.2022.12808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/14/2021] [Indexed: 12/04/2022] Open
Abstract
Osteoporosis (OP) is a bone metabolic disease, in which low bone mass and the microarchitectural deterioration of bone tissue contribute to the fragility of bones and increase the risk of fracture. The aim of the present study was to determine the role of microRNA (miR)-98-5p in high glucose (HG)-induced preosteoblasts. HG was used to induce preosteoblasts treated in a differentiation medium to establish an in vitro OP model. Next, miR-98-5p expression was determined using reverse transcription-quantitative PCR. Following the transfection of an miR-98-5p inhibitor into HG-treated osteoblasts, cell viability was assessed using a Cell Counting Kit-8 assay, while alkaline phosphatase (ALP) activity, differentiation ability and the expression of differentiation-regulated genes osteocalcin and osteopontin were measured using the corresponding ALP, Alizarin red staining, reverse transcription-quantitative PCR and western blotting assays. The association between miR-98-5p and the PI3K/AKT/GSK3β signaling pathway was determined using western blotting. Next, the binding relationship between miR-98-5p and bone morphogenetic protein 2 (BMP2) was predicted and verified, and the role of BMP2 in the regulation of the PI3K/AKT/GSK3β signaling pathway was explored using western blotting. The results revealed that miR-98-5p expression was upregulated in HG-induced osteoblasts, and the inhibition of miR-98-5p resulted in enhanced cell viability, alkaline phosphatase activity and differentiation in osteoblasts following HG induction. It was also discovered that miR-98-5p inhibition activated PI3K/AKT/GSK3β signaling, while knockdown of BMP2, which binds to miR-98-5p, enhanced the activation of this signaling pathway and the differentiation ability of osteoblasts. In conclusion, the findings of the present study suggested that the inhibition of miR-98-5p expression may activate PI3K/AKT/GSK3β signaling to promote HG-induced suppression of preosteoblast viability and differentiation by targeting BMP2, which provides a novel insight into future potential molecular markers for OP treatment.
Collapse
Affiliation(s)
- Feng Zheng
- Department of Orthopedics, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Fucai Zhang
- Department of Orthopedics, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Furong Wang
- Department of Orthopedics, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| |
Collapse
|
26
|
Malakoti F, Zare F, Zarezadeh R, Raei Sadigh A, Sadeghpour A, Majidinia M, Yousefi B, Alemi F. The role of melatonin in bone regeneration: A review of involved signaling pathways. Biochimie 2022; 202:56-70. [PMID: 36007758 DOI: 10.1016/j.biochi.2022.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/27/2022] [Accepted: 08/11/2022] [Indexed: 11/29/2022]
Abstract
Increasing bone resorption followed by decreasing bone mineralization are hallmarks of bone degeneration, which mostly occurs in the elderly population and post-menopausal women. The use of mesenchymal stem cells (MSCs) has raised many promises in the field of bone regeneration due to their high osteoblastic differentiation capacity and easy availability from abundant sources. A variety of compounds, including growth factors, cytokines, and other internal factors, have been combined with MSCs to increase their osteoblastic differentiation capacity. One of these factors is melatonin, whose possible regulatory role in bone metabolism and formation has recently been suggested by many studies. Melatonin also is a potential signaling molecule and can affect many of the signaling pathways involved in MSCs osteoblastic differentiation, such as activation of PI3K/AKT, BMP/Smad, MAPK, NFkB, Nrf2/HO-1, Wnt, SIRT/SOD, PERK/ATF4. Furthermore, melatonin in combination with other components such as strontium, vitamin D3, and vitamin K2 has a synergistic effect on bone microstructure and improves bone mineral density (BMD). In this review article, we aim to summarize the regulatory mechanisms of melatonin in osteoblastic differentiation of MSCs and underling involved signaling pathways as well as the clinical potential of using melatonin in bone degenerative disorders.
Collapse
Affiliation(s)
- Faezeh Malakoti
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farshad Zare
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aydin Raei Sadigh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Sadeghpour
- Department of Orthopedic Surgery, School of Medicine and Shohada Educational Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Li M, Yang N, Hao L, Zhou W, Li L, Liu L, Yang F, Xu L, Yao G, Zhu C, Xu W, Fang S. Melatonin Inhibits the Ferroptosis Pathway in Rat Bone Marrow Mesenchymal Stem Cells by Activating the PI3K/AKT/mTOR Signaling Axis to Attenuate Steroid-Induced Osteoporosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8223737. [PMID: 36035224 PMCID: PMC9410838 DOI: 10.1155/2022/8223737] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 12/12/2022]
Abstract
Steroid-induced osteoporosis (SIOP) is a form of secondary osteoporosis, but its specific mechanism remains unclear. Glucocorticoid (GC-)-induced death of osteoblasts and bone marrow mesenchymal stem cells (BMSCs) is an important factor in SIOP. Ferroptosis is an iron-dependent type of programmed cell death and can be induced by many factors. Herein, we aimed to explore whether GCs cause ferroptosis of BMSCs, identify pathways as possible therapeutic targets, and determine the underlying mechanisms of action. In this study, we used high-dose dexamethasone (DEX) to observe whether GCs induce ferroptosis of BMSCs. Additionally, we established a rat SIOP model and then assessed whether melatonin (MT) could inhibit the ferroptosis pathway to provide early protection against GC-induced SIOP and investigated the signaling pathways involved. In vitro experiments confirmed that DEX induces ferroptosis in BMSCs. MT significantly alleviates GC-induced ferroptosis of BMSCs. Pathway analysis showed that MT ameliorates ferroptosis by activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) axis. MT upregulates the expression of PI3K, which is an important regulator of ferroptosis resistance. PI3K activators mimic the antiferroptotic effect of MT, but when the PI3K pathway is blocked, the effect of MT is weakened. Using in vivo experiments, we confirmed the in vitro results and observed that MT can obviously protect against SIOP induced by GC. Notably, even after the initiation of GC-induced ferroptosis, MT can confer protection against SIOP. Our research confirms that GC-induced ferroptosis is closely related to SIOP. MT can inhibit ferroptosis by activating the PI3K/AKT/mTOR signaling pathway, thereby inhibiting the occurrence of SIOP. Therefore, MT may be a novel agent for preventing and treating SIOP.
Collapse
Affiliation(s)
- Meng Li
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Ning Yang
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Li Hao
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Wei Zhou
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Lei Li
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Lei Liu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Fang Yang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
| | - Lei Xu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Gang Yao
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Chen Zhu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Wei Xu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| | - Shiyuan Fang
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road No. 17, Hefei, 230001 Anhui, China
| |
Collapse
|
28
|
Zhao Y, Shao G, Liu X, Li Z. Assessment of the Therapeutic Potential of Melatonin for the Treatment of Osteoporosis Through a Narrative Review of Its Signaling and Preclinical and Clinical Studies. Front Pharmacol 2022; 13:866625. [PMID: 35645810 PMCID: PMC9130700 DOI: 10.3389/fphar.2022.866625] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/06/2022] [Indexed: 12/21/2022] Open
Abstract
Melatonin is a bioamine produced primarily in the pineal gland, although peripheral sites, including the gut, may also be its minor source. Melatonin regulates various functions, including circadian rhythm, reproduction, temperature regulation, immune system, cardiovascular system, energy metabolism, and bone metabolism. Studies on cultured bone cells, preclinical disease models of bone loss, and clinical trials suggest favorable modulation of bone metabolism by melatonin. This narrative review gives a comprehensive account of the current understanding of melatonin at the cell/molecular to the systems levels. Melatonin predominantly acts through its cognate receptors, of which melatonin receptor 2 (MT2R) is expressed in mesenchymal stem cells (MSCs), osteoblasts (bone-forming), and osteoclasts (bone-resorbing). Melatonin favors the osteoblastic fate of MSCs, stimulates osteoblast survival and differentiation, and inhibits osteoclastogenic differentiation of hematopoietic stem cells. Produced from osteoblastic cells, osteoprotegerin (OPG) and receptor activator of nuclear factor kappa B ligand (RANKL) critically regulate osteoclastogenesis and melatonin by suppressing the osteoclastogenic RANKL, and upregulating the anti-osteoclastogenic OPG exerts a strong anti-resorptive effect. Although the anti-inflammatory role of melatonin favors osteogenic function and antagonizes the osteoclastogenic function with the participation of SIRT signaling, various miRNAs also mediate the effects of the hormone on bone cells. In rodent models of osteoporosis, melatonin has been unequivocally shown to have an anti-osteoporotic effect. Several clinical trials indicate the bone mass conserving effect of melatonin in aging/postmenopausal osteoporosis. This review aims to determine the possibility of melatonin as a novel class of anti-osteoporosis therapy through the critical assessment of the available literature.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Guoxi Shao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Xingang Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Zhengwei Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
29
|
Wang Z, Chen T, Wu Z, Jiang X, Hou Q, Miao S, Xia R, Wang L. The dual-effects of PLGA@MT electrospun nanofiber coatings on promoting osteogenesis at the titanium-bone interface under diabetic conditions. J Mater Chem B 2022; 10:4020-4030. [PMID: 35506736 DOI: 10.1039/d2tb00120a] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The high failure risk of endosseous titanium implants under diabetes conditions appeals to strengthen the osteointegration on the titanium-bone (Ti-B) interface. Melatonin (MT) is a neurohormone involved in bone homeostasis, which can promote osteogenesis and inhibit ROS overproduction through multiple pathways, but its effects on the Ti-B interface in diabetes remain elusive. The biodegradable poly(lactic-co-glycolic acid) (PLGA) has excellent controlled and sustained release properties, low cytotoxicity, and biocompatibility. Our study fabricated a nanofiber in which MT was encapsulated in PLGA to generate a nanofiber coating on a polydopamine (PDA)-modified titanium surface using electrospinning technology. The surface characteristic showed that MT was fully encapsulated in the PLGA carrier, and PLGA@MT was strongly coupled to the titanium matrix. Furthermore, the PLGA@MT-Ti nanofiber could release MT for at least 30 days. In vitro cellular tests demonstrated that PLGA@MT-Ti directly stimulates osteogenesis on the Ti-B interface by activating the BMP-4/WNT pathway in a dose-dependent manner. The effect of suppressing diabetes-induced ROS overproduction and promoting cell proliferation was not proportional to the content of MT. In vivo experiments revealed that PLGA@MT-Ti screws promoted the bone formation and osteointegration in type 1 diabetes mellitus (T1DM) mice with tibial bone defects. Our findings demonstrate that PLGA@MT-Ti exerted dual effects through activating the BMP-4/WNT pathway and attenuating ROS overproduction to promote osteogenesis and osteointegration at the Ti-B interface, providing a novel strategy to fabricate biomaterial modification and biofunctionalization under diabetic conditions.
Collapse
Affiliation(s)
- Zijie Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Tingting Chen
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Zimei Wu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xingzhu Jiang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Qiaodan Hou
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Sikai Miao
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Ruihao Xia
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Lin Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China. .,Southern University of Science and Technology Hospital, Shenzhen, 518055, China
| |
Collapse
|
30
|
McCarty MF, Lewis Lujan L, Iloki Assanga S. Targeting Sirt1, AMPK, Nrf2, CK2, and Soluble Guanylate Cyclase with Nutraceuticals: A Practical Strategy for Preserving Bone Mass. Int J Mol Sci 2022; 23:4776. [PMID: 35563167 PMCID: PMC9104509 DOI: 10.3390/ijms23094776] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
There is a vast pre-clinical literature suggesting that certain nutraceuticals have the potential to aid the preservation of bone mass in the context of estrogen withdrawal, glucocorticoid treatment, chronic inflammation, or aging. In an effort to bring some logical clarity to these findings, the signaling pathways regulating osteoblast, osteocyte, and osteoclast induction, activity, and survival are briefly reviewed in the present study. The focus is placed on the following factors: the mechanisms that induce and activate the RUNX2 transcription factor, a key driver of osteoblast differentiation and function; the promotion of autophagy and prevention of apoptosis in osteoblasts/osteoclasts; and the induction and activation of NFATc1, which promotes the expression of many proteins required for osteoclast-mediated osteolysis. This analysis suggests that the activation of sirtuin 1 (Sirt1), AMP-activated protein kinase (AMPK), the Nrf2 transcription factor, and soluble guanylate cyclase (sGC) can be expected to aid the maintenance of bone mass, whereas the inhibition of the serine kinase CK2 should also be protective in this regard. Fortuitously, nutraceuticals are available to address each of these targets. Sirt1 activation can be promoted with ferulic acid, N1-methylnicotinamide, melatonin, nicotinamide riboside, glucosamine, and thymoquinone. Berberine, such as the drug metformin, is a clinically useful activator of AMPK. Many agents, including lipoic acid, melatonin, thymoquinone, astaxanthin, and crucifera-derived sulforaphane, can promote Nrf2 activity. Pharmacological doses of biotin can directly stimulate sGC. Additionally, certain flavonols, notably quercetin, can inhibit CK2 in high nanomolar concentrations that may be clinically relevant. Many, though not all, of these agents have shown favorable effects on bone density and structure in rodent models of bone loss. Complex nutraceutical regimens providing a selection of these nutraceuticals in clinically meaningful doses may have an important potential for preserving bone health. Concurrent supplementation with taurine, N-acetylcysteine, vitamins D and K2, and minerals, including magnesium, zinc, and manganese, plus a diet naturally high in potassium, may also be helpful in this regard.
Collapse
Affiliation(s)
| | - Lidianys Lewis Lujan
- Department of Research and Postgraduate in Food Science, Sonoran University, Hermosillo 83200, Mexico;
| | - Simon Iloki Assanga
- Department of Biological Chemical Sciences, Sonoran University, Hermosillo 83200, Mexico;
| |
Collapse
|
31
|
Cao L, Yang K, Yuan W, Zhou S, Zhao R, Qiu S. Melatonin Mediates Osteoblast Proliferation Through the STIM1/ORAI1 Pathway. Front Pharmacol 2022; 13:851663. [PMID: 35392575 PMCID: PMC8980543 DOI: 10.3389/fphar.2022.851663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/08/2022] [Indexed: 01/09/2023] Open
Abstract
Based on the positive correlation between bone mineral density and melatonin levels in blood, this study confirmed that melatonin supplementation prevents postmenopausal osteoporosis. We further confirmed that melatonin promotes an increase in intracellular calcium concentrations through the STIM1/ORAI1 pathway, thereby inducing the proliferation of osteoblasts. Introduction: Osteoporosis (OP) is a progressive, systemic bone disease that is one of the main causes of disability and death in elderly female patients. As an amine hormone produced by the human pineal gland, melatonin plays an important role in regulating bone metabolism. This study intends to investigate the relationship between melatonin levels in human blood and bone density and to suggest the efficacy of melatonin in treating osteoporosis by performing in vivo and in vitro experiments. Methods: We used liquid chromatography-tandem mass spectrometry to determine the serum melatonin levels in postmenopausal women with osteoporosis and young women with a normal bone mass. The bone density, BV/TV, Tb.Th, Tb.Sp and other indicators of postmenopausal osteoporosis and mice with a normal bone mass were detected by measuring bone density and micro-CT. The intracellular calcium ion concentration was detected using fluorescence microscopy and a full-wavelength multifunctional microplate reader, and the expression of SOCE-related genes and STIM1/ORAI1 proteins was detected using PCR and WB. Results: This study confirmed that bone density positively correlates with the melatonin level in human blood. In the animal model, melatonin supplementation reverses postmenopausal osteoporosis. We explored the internal mechanism of melatonin treatment of osteoporosis. Melatonin promotes an increase in intracellular calcium ion concentrations through the STIM1/ORAI1 pathway to induce osteoblast proliferation. Conclusions: This study provides an important theoretical basis for the clinical application of melatonin in patients with osteoporosis and helps to optimize the diagnosis and treatment of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Lili Cao
- Department of Medical Oncology, First Hospital of China Medical University, Shenyang, China
| | - Keda Yang
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
| | - Wei Yuan
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
| | - Siming Zhou
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
| | - Rui Zhao
- Minimally Invasive Orthopedic Surgery Ward, Shenyang Orthopedic Hospital, Shenyang, China
| | - Shui Qiu
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
32
|
Yang K, Cao F, Xue Y, Tao L, Zhu Y. Three Classes of Antioxidant Defense Systems and the Development of Postmenopausal Osteoporosis. Front Physiol 2022; 13:840293. [PMID: 35309045 PMCID: PMC8927967 DOI: 10.3389/fphys.2022.840293] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 01/04/2023] Open
Abstract
Osteoporosis is a common bone imbalance disease that threatens the health of postmenopausal women. Estrogen deficiency accelerates the aging of women. Oxidative stress damage is regarded as the main pathogenesis of postmenopausal osteoporosis. The accumulation of reactive oxygen species in the bone microenvironment plays a role in osteoblast and osteoclast apoptosis. Improving the oxidative state is essential for the prevention and treatment of postmenopausal osteoporosis. There are three classes of antioxidant defense systems in the body to eliminate free radicals and peroxides including antioxidant substances, antioxidant enzymes, and repair enzymes. In our review, we demonstrated the mechanism of antioxidants and their effect on bone metabolism in detail. We concluded that glutathione/oxidized glutathione (GSH/GSSG) conversion involved the PI3K/Akt-Nrf2/HO-1 signaling pathway and that the antioxidant enzyme-mediated mitochondrial apoptosis pathway of osteoblasts was necessary for the development of postmenopausal osteoporosis. Since the current therapeutic effects of targeting bone cells are not significant, improving the systemic peroxidation state and then regulating bone homeostasis will be a new method for the treatment of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Keda Yang
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
| | - Fangming Cao
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
| | - Yuchuan Xue
- The First Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
- *Correspondence: Lin Tao,
| | - Yue Zhu
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
- Yue Zhu,
| |
Collapse
|
33
|
Zhang F, Li Q, Wu J, Ruan H, Sun C, Zhu J, Song Q, Wei X, Shi Y, Zhu L. Total Flavonoids of Drynariae Rhizoma Improve Glucocorticoid-Induced Osteoporosis of Rats: UHPLC-MS-Based Qualitative Analysis, Network Pharmacology Strategy and Pharmacodynamic Validation. Front Endocrinol (Lausanne) 2022; 13:920931. [PMID: 35846330 PMCID: PMC9279576 DOI: 10.3389/fendo.2022.920931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Glucocorticoid-induced osteoporosis (GIOP) is a common form of secondary osteoporosis caused by the protracted or a large dosage of glucocorticoids (GCs). Total flavonoids of Drynariae rhizoma (TFDR) have been widely used in treating postmenopausal osteoporosis (POP). However, their therapeutic effects and potential mechanism against GIOP have not been fully elucidated. METHODS Ultra-high-performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight mass spectrometry (UHPLC-ESIQ-TOF-MS) experiments were performed for qualitative analysis. We performed hematoxylin-eosin (HE) staining and microcomputed tomography (micro-CT) analysis to detect the changes in bone microstructure. The changes in biochemical parameters in the serum samples were determined by performing an enzyme-linked immunosorbent assay (ELISA). The prediction results of network pharmacology were verified via quantitative real-time polymerase chain reaction (qRT-PCR) to elucidate the potential mechanism of TFDR against GIOP. RESULTS A total of 191 ingredients were identified in vitro and 48 ingredients in vivo. In the in-vivo experiment, the levels of the serum total cholesterol (TC), the serum triglyceride (TG), Leptin (LEP), osteocalcin (OC), osteoprotegerin (OPG), bone morphogenetic protein-2 (BMP-2), propeptide of type I procollagen (PINP), tartrate-resistant acid phosphatase (TRACP) and type-I collagen carboxy-terminal peptide (CTX-1) in the TFDR group significantly changed compared with those in the GIOP group. Moreover, the TFDR group showed an improvement in bone mineral density and bone microstructure. Based on the results of network pharmacology analysis, 67 core targets were selected to construct the network and perform PPI analysis as well as biological enrichment analysis. Five of the targets with high "degree value" had differential gene expression between groups using qRT-PCR. CONCLUSION TFDR, which may play a crucial role between adipose metabolism and bone metabolism, may be a novel remedy for the prevention and clinical treatment of GIOP.
Collapse
Affiliation(s)
- Fangqing Zhang
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiuyue Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiashuo Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haonan Ruan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuanrui Sun
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jia Zhu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qinghui Song
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xu Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Xu Wei, ; Yue Shi, ; Liguo Zhu,
| | - Yue Shi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Xu Wei, ; Yue Shi, ; Liguo Zhu,
| | - Liguo Zhu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Xu Wei, ; Yue Shi, ; Liguo Zhu,
| |
Collapse
|
34
|
Han L, Liu H, Fu H, Hu Y, Fang W, Liu J. Exosome-delivered BMP-2 and polyaspartic acid promotes tendon bone healing in rotator cuff tear via Smad/RUNX2 signaling pathway. Bioengineered 2022; 13:1459-1475. [PMID: 35258414 PMCID: PMC8805918 DOI: 10.1080/21655979.2021.2019871] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/13/2021] [Indexed: 11/02/2022] Open
Abstract
Rotator cuff tear is the main form of shoulder joint injury, which seriously affects shoulder joint function. This study aimed to clarify the function and mechanism of exosomes containing polylactic acid (PLA), polylactic acid copolymer and BMP-2 in tendon bone healing of rotator cuff tear. First, CD44 expression in bone marrow mesenchymal stem cells (BMSCs) and CD90 and CD44 in exosomes were analyzed by flow cytometry. Then, stability and targeting identification of exosome-delivered bone morphogenetic protein (BMP)-2 and PLA microcapsules were measured by transmission electron microscopy (TEM), DiO/DiI staining. Finally, tendon-bone repair after acute rotator cuff rupture in rabbits was established, and the function of BMP-2 exosomes for tendon bone healing in rotator cuff tear was evaluated by micro-CT, biomechanical determination and histochemical staining methods. The results showed that the exosomes of polyaspartic acid-polylactic acid-glycolic acid copolymer (PASP-PLGA) microcapsules were successfully established which showed good stability and targeting. The bone mineral density (BMD), tissue mineral density (TMD) and bone volume fraction (BV/TV) were higher, while the stiffness and the ultimate load strength of the tendon interface were enhanced under treatment with exosomes of PASP-PLGA microcapsules. Histochemical staining showed that exosomes of PASP-PLGA microcapsules promoted tendon and bone interface healing after rotator cuff injury. The tendon regeneration- and cartilage differentiation-related protein expressions were significantly upregulated under treatment with exosomes of PASP-PLGA microcapsules. In conclusion, exosome-delivered BMP-2 and PLA promoted tendon bone healing in rotator cuff tear via Smad/RUNX2 pathway. Our findings may provide a new insight for promoting tendon healing.
Collapse
Affiliation(s)
- Lei Han
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Hong Liu
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Huajun Fu
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Yugen Hu
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Weili Fang
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Junsheng Liu
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Wang Q, Xie X, Zhang D, Mao F, Wang S, Liao Y. Saxagliptin enhances osteogenic differentiation in MC3T3-E1 cells, dependent on the activation of AMP-activated protein kinase α (AMPKα)/runt-related transcription factor-2 (Runx-2). Bioengineered 2022; 13:431-439. [PMID: 35258398 PMCID: PMC8805826 DOI: 10.1080/21655979.2021.2008667] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/16/2021] [Indexed: 12/29/2022] Open
Abstract
Osteoporosis is a metabolic bone disease commonly observed in the elderly, and its pathogenesis is associated with declined osteogenic differentiation. Osteogenic differentiation could be facilitated by the activation of the AMP-activated protein kinase (AMPK) pathway. Saxagliptin, an anti-diabetic agent with inhibitory effects against dipeptidyl peptidase 4 (DPP-4), has been recently reported to induce the activation of the AMPK pathway. The present study proposes to explore the function and mechanism of Saxagliptin in osteogenic differentiation. Osteogenic differentiation induction medium (ODIM) was utilized to induce osteogenic differentiation in MC3T3-E1 cells. Significantly increased mineral nodule formation, elevated alkaline phosphatase (ALP) activity, and upregulated expression of osteogenic marker genes activating transcription factor-4 (ATF-4), osteopontin (OPN), and type I collagen (Col1) were observed in ODIM-cultured MC3T3-E1 cells, all of which were further enhanced by the introduction of Saxagliptin. The elevated expression level of runt-related transcription factor-2 (Runx-2), an important transcriptional factor involved in the progression of osteogenic differentiation, in ODIM-cultured MC3T3-E1 cells was further promoted by Saxagliptin. The AMPK pathway in ODIM-cultured MC3T3-E1 cells was significantly activated by Saxagliptin, and the functions of Saxagliptin in promoting osteogenic differentiation were abolished by compound C, the inhibitor of the AMPK pathway. Conclusively, Saxagliptin enhanced osteogenic differentiation in MC3T3-E1 cells, dependent on the activation of AMPKα/RUNX-2.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Orthopaedics, The 5th People’s Hospital of Shanghai Fudan University, Shanghai, China
| | - Xiaoxing Xie
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Dehua Zhang
- Department of Orthopaedics, The Central Hospital of Karamay, Xinjiang, Karamay, Xinjiang, China
| | - Feng Mao
- Department of Orthopaedics, The Central Hospital of Karamay, Xinjiang, Karamay, Xinjiang, China
| | - Shaobo Wang
- Department of Orthopaedics, The Central Hospital of Karamay, Xinjiang, Karamay, Xinjiang, China
| | - Yi Liao
- Department of Orthopaedics, The Central Hospital of Karamay, Xinjiang, Karamay, Xinjiang, China
| |
Collapse
|
36
|
Feng ZY, Yang SD, Wang T, Guo S. Effect of Melatonin for Regulating Mesenchymal Stromal Cells and Derived Extracellular Vesicles. Front Cell Dev Biol 2021; 9:717913. [PMID: 34540834 PMCID: PMC8440901 DOI: 10.3389/fcell.2021.717913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
Melatonin is a hormone, synthesized in the pineal gland, which primarily controls the circadian rhythm of the body. In recent years, melatonin has also been shown to regulate metabolism, provide neuroprotection, and act as an anti-inflammatory, free radical scavenger. There has also been a recent research interest in the role of melatonin in regulating mesenchymal stromal cells (MSCs). MSCs are pivotal for their ability to differentiate into a variety of different tissues. There is also increasing evidence for the therapeutic prospects of MSCs via paracrine signaling. In addition to secreting cytokines and chemokines, MSCs can secrete extracellular vesicles (EVs), allowing them to respond to injury and promote tissue regeneration. While there has been a major research interest in the use of MSCs for regenerative medicine, the clinical application is limited by many risks, including tumorigenicity, senescence, and sensitivity to toxic environments. The use of MSC-derived EVs for cell-free therapy can potentially avoid the disadvantages of MSCs, which makes this an exciting prospect for regenerative medicine. Prior research has shown that MSCs, via paracrine mechanisms, can identify receptor-independent responses to melatonin and then activate a series of downstream pathways, which exert a variety of effects, including anti-tumor and anti-inflammatory effects. Here we review the synthesis of melatonin, its mechanisms of action, and the effect of melatonin on MSCs via paracrine signaling. Furthermore, we summarize the current clinical applications of melatonin and discuss future prospects.
Collapse
Affiliation(s)
- Zi-Yi Feng
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shu-De Yang
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ting Wang
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
37
|
MacDonald IJ, Tsai HC, Chang AC, Huang CC, Yang SF, Tang CH. Melatonin Inhibits Osteoclastogenesis and Osteolytic Bone Metastasis: Implications for Osteoporosis. Int J Mol Sci 2021; 22:ijms22179435. [PMID: 34502344 PMCID: PMC8430520 DOI: 10.3390/ijms22179435] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/16/2022] Open
Abstract
Osteoblasts and osteoclasts are major cellular components in the bone microenvironment and they play a key role in the bone turnover cycle. Many risk factors interfere with this cycle and contribute to bone-wasting diseases that progressively destroy bone and markedly reduce quality of life. Melatonin (N-acetyl-5-methoxy-tryptamine) has demonstrated intriguing therapeutic potential in the bone microenvironment, with reported effects that include the regulation of bone metabolism, acceleration of osteoblastogenesis, inhibition of osteoclastogenesis and the induction of apoptosis in mature osteoclasts, as well as the suppression of osteolytic bone metastasis. This review aims to shed light on molecular and clinical evidence that points to possibilities of melatonin for the treatment of both osteoporosis and osteolytic bone metastasis. It appears that the therapeutic qualities of melatonin supplementation may enable existing antiresorptive osteoporotic drugs to treat osteolytic metastasis.
Collapse
Affiliation(s)
- Iona J. MacDonald
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan; (I.J.M.); (H.-C.T.)
| | - Hsiao-Chi Tsai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan; (I.J.M.); (H.-C.T.)
| | - An-Chen Chang
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei City 111, Taiwan;
| | - Chien-Chung Huang
- School of Medicine, China Medical University, Taichung 40402, Taiwan;
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung 40447, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan; (I.J.M.); (H.-C.T.)
- School of Medicine, China Medical University, Taichung 40402, Taiwan;
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung 41354, Taiwan
- Correspondence: ; Tel.: +886-2205-2121 (ext. 7726)
| |
Collapse
|
38
|
Povoroznyuk VV, Dedukh NV, Bystrytska MA, Shapovalov VS. Bone remodeling stages under physiological conditions and glucocorticoid in excess: Focus on cellular and molecular mechanisms. REGULATORY MECHANISMS IN BIOSYSTEMS 2021. [DOI: 10.15421/022130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
This review provides a rationale for the cellular and molecular mechanisms of bone remodeling stages under physiological conditions and glucocorticoids (GCs) in excess. Remodeling is a synchronous process involving bone resorption and formation, proceeding through stages of: (1) resting bone, (2) activation, (3) bone resorption, (4) reversal, (5) formation, (6) termination. Bone remodeling is strictly controlled by local and systemic regulatory signaling molecules. This review presents current data on the interaction of osteoclasts, osteoblasts and osteocytes in bone remodeling and defines the role of osteoprogenitor cells located above the resorption area in the form of canopies and populating resorption cavities. The signaling pathways of proliferation, differentiation, viability, and cell death during remodeling are presented. The study of signaling pathways is critical to understanding bone remodeling under normal and pathological conditions. The main signaling pathways that control bone resorption and formation are RANK / RANKL / OPG; M-CSF – c-FMS; canonical and non-canonical signaling pathways Wnt; Notch; MARK; TGFβ / SMAD; ephrinB1/ephrinB2 – EphB4, TNFα – TNFβ, and Bim – Bax/Bak. Cytokines, growth factors, prostaglandins, parathyroid hormone, vitamin D, calcitonin, and estrogens also act as regulators of bone remodeling. The role of non-encoding microRNAs and long RNAs in the process of bone cell differentiation has been established. MicroRNAs affect many target genes, have both a repressive effect on bone formation and activate osteoblast differentiation in different ways. Excess of glucocorticoids negatively affects all stages of bone remodeling, disrupts molecular signaling, induces apoptosis of osteocytes and osteoblasts in different ways, and increases the life cycle of osteoclasts. Glucocorticoids disrupt the reversal stage, which is critical for the subsequent stages of remodeling. Negative effects of GCs on signaling molecules of the canonical Wingless (WNT)/β-catenin pathway and other signaling pathways impair osteoblastogenesis. Under the influence of excess glucocorticoids biosynthesis of biologically active growth factors is reduced, which leads to a decrease in the expression by osteoblasts of molecules that form the osteoid. Glucocorticoids stimulate the expression of mineralization inhibitor proteins, osteoid mineralization is delayed, which is accompanied by increased local matrix demineralization. Although many signaling pathways involved in bone resorption and formation have been discovered and described, the temporal and spatial mechanisms of their sequential turn-on and turn-off in cell proliferation and differentiation require additional research.
Collapse
|
39
|
Polo Like Kinase 4 (PLK4) impairs human bone marrow mesenchymal stem cell (BMSC) viability and osteogenic differentiation. Biochem Biophys Res Commun 2021; 549:221-228. [PMID: 33706192 DOI: 10.1016/j.bbrc.2021.02.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/07/2021] [Indexed: 12/12/2022]
Abstract
Human bone marrow mesenchymal stem cell (hBMSC) viability and osteogenic differentiation play a critical role in bone disorders such as osteoporosis. In the present study, we identified the aberrant PLK4 upregulation in osteoporosis and downregulation in BMSCs during osteogenic differentiation. In isolated hBMSCs, PLK4 overexpression significantly inhibited, whereas PLK4 knockdown promoted cell viability and hBMSC osteogenic differentiation. For molecular mechanism, PLK4 overexpression decreased, whereas PLK4 knockdown increased WNT1 and β-catenin protein levels and the phosphorylation of Smad1/5/8. The Wnt/β-catenin signaling antagonist Dickkopf 1 (DKK1) or the BMP-Smads antagonist LDN193189 dramatically suppressed hBMSC osteoblast differentiation, and partially attenuated the promotive effects of PLK4 knockdown on hBMSC osteogenic differentiation. Altogether, PLK4 overexpression impairs hBMSC viability and osteogenic differentiation potential, possibly through the Wnt/β-catenin signaling and BMP/Smads signaling.
Collapse
|
40
|
Bai Y, Zhang Q, Chen Q, Zhou Q, Zhang Y, Shi Z, Nong H, Liu M, Zeng G, Zong S. Conditional knockout of the PDK-1 gene in osteoblasts affects osteoblast differentiation and bone formation. J Cell Physiol 2020; 236:5432-5445. [PMID: 33377210 DOI: 10.1002/jcp.30249] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022]
Abstract
Osteoblasts are the main functional cells of bone formation, and they are responsible for the synthesis, secretion, and mineralization of the bone matrix. Phosphatidylinositol-3-kinase/Akt is an important signaling pathway involved in the regulation of cell proliferation, death, and survival. Some studies have shown that 3-phosphoinositide-dependent protein kinase-1 (PDK-1) plays an important role in the phosphorylation of Akt. In the present study, an osteocalcin (OCN) promoter-driven Cre-LoxP system was established to specifically delete the PDK-1 gene in osteoblasts. It was found that the size and weight of PDK-1 conditional gene knockout (cKO) mice were significantly reduced. von Kossa staining and microcomputed tomography showed that the trabecular thickness, trabecular number, and bone volume were significantly decreased, whereas trabecular separation was increased, as compared with wide-type littermates, which were characterized by a decreased bone mass. A model of distal femoral defect was established, and it was found that cKO mice delayed bone defect repair. In osteoblasts derived from PDK-1 cKO mice, the alkaline phosphatase (ALP) secretion and ability of calcium mineralization were significantly decreased, and the expressions of osteoblast-related proteins, runt-related transcription factor 2, OCN, and ALP were also clearly decreased. Moreover, the phosphorylation level of Akt and downstream factor GSK3β and their response to insulin-like growth factor-1 (IGF-1) decreased clearly. Therefore, we believe that PDK-1 plays a very important role in osteoblast differentiation and bone formation by regulating the PDK-1/Akt/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Yiguang Bai
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.,Department of Orthopaedics, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qiong Zhang
- Department of Nutrition and Food Hygiene, College of Public Hygiene of Guangxi Medical University, Nanning, Guangxi, China
| | - Qiaoling Chen
- Department of Oncology, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Quan Zhou
- Collaborative Innovation Center of Guangxi Biological Medicine, Guangxi Medical University, Nanning, Guangxi, China.,Department of Emergency, The Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Yanan Zhang
- Collaborative Innovation Center of Guangxi Biological Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhuohua Shi
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Haibin Nong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Mingfu Liu
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Gaofeng Zeng
- Department of Nutrition and Food Hygiene, College of Public Hygiene of Guangxi Medical University, Nanning, Guangxi, China
| | - Shaohui Zong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.,Research Centre for Regenerative Medicine and Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|