1
|
Shi K, Wei J, Chen J. MiR-223-3p Promotes Osteoporosis Progression by Repressing Osteogenic Differentiation via Targeting FHL1/Wnt/β-catenin Signaling. Cell Biochem Biophys 2025; 83:1703-1711. [PMID: 39613991 DOI: 10.1007/s12013-024-01579-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 12/01/2024]
Abstract
The aim of this research was to unveil the potential along with potential mechanism of miR-223-3p in osteoporosis. RT-qPCR together with western blot was implemented to examine miR-223-3p, FHL1, along with osteogenic markers levels during bone marrow mesenchymal stem cells (BMSCs) differentiation. The ALP activity staining along with alizarin red staining (ARS) were implemented to assess ALP activity as well as the mineralization ability of BMSCs. Binding sequences for miR-223-3p and FHL1 from starBase website were validated through dual-luciferase reporter gene assay. MiR-223-3p was down-regulated in BMSCs during osteoblasts differentiation, and miR-223-3p elevation hindered BMSCs' osteogenic differentiation. FHL1 belonged to the target mRNA of miR-223-3p. FHL1 presented up-regulation in BMSCs during osteoblasts differentiation. More importantly, FHL1 expression was negative modulated by miR-223-3p in BMSCs during osteoblasts differentiation, and FHL1 elevation could inverse the inhibited BMSCs' osteogenic differentiation modulated by miR-223-3p elevation. Furthermore, miR-223-3p elevation repressed the Wnt/β-catenin pathway activity in lithium chloride-treated BMSCs, and FHL1 overexpression counteracted the inhibitory effect of the Wnt/β-catenin pathway caused by miR-223-3p up-regulation. Collectively, miR-223-3p accelerates osteoporosis progression by repressing osteogenic differentiation through targeting FHL1/Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Kairi Shi
- Department of Orthopedic Surgery, Ningbo No.6 Hospital, Ningbo, Zhejiang, 315040, China
| | - Junyu Wei
- Department of Orthopedic Surgery, Ningbo No.6 Hospital, Ningbo, Zhejiang, 315040, China
| | - Jianming Chen
- Department of Orthopedic Surgery, Ningbo No.6 Hospital, Ningbo, Zhejiang, 315040, China.
| |
Collapse
|
2
|
Wen L, Ye R, Zhai W, Li D, Sun H. Efferocytosis in inflammatory bone disorders. Trends Pharmacol Sci 2025:S0165-6147(25)00067-7. [PMID: 40348687 DOI: 10.1016/j.tips.2025.04.001] [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: 12/03/2024] [Revised: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 05/14/2025]
Abstract
Efferocytosis, the clearance of apoptotic cells (ACs) by phagocytes, is crucial for bone homeostasis and immune balance. This tightly regulated process depends on molecular markers such as phosphatidylserine on ACs and MERTK on phagocytes. In the bone microenvironment, multiple cell types participate in efferocytosis, including osteal macrophages, mesenchymal stem cells, osteoblasts, and osteoclasts, directly influencing bone remodeling and immune responses. Impaired efferocytosis disrupts bone turnover, exacerbates inflammation, and contributes to inflammatory bone diseases. Despite its recognized importance, the precise mechanisms regulating efferocytosis in osteoimmunology remain underexplored, including specific signaling pathways, cell-specific interactions, and therapeutic applications. Recent advances highlight the therapeutic potential of targeting efferocytosis using modalities and biomaterial-based strategies. This review systematically examines the role of efferocytosis in osteoimmunology, discusses key challenges in its therapeutic translation, and explores emerging strategies to optimize efferocytosis-based interventions for inflammatory bone disorders.
Collapse
Affiliation(s)
- Linlin Wen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, China
| | - Rongrong Ye
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, China
| | - Wenhao Zhai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, China.
| | - Hongchen Sun
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, China.
| |
Collapse
|
3
|
Teng Z, Zhu J, Li K, Tong T, Li W, Chu H, Sun P. Efficacy and safety of acupuncture as an adjuvant therapy for osteoporosis: a systematic review and meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne) 2025; 16:1561344. [PMID: 40416525 PMCID: PMC12098033 DOI: 10.3389/fendo.2025.1561344] [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: 01/15/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025] Open
Abstract
Objective To systematically evaluate the efficacy and safety of acupuncture as an adjuvant therapy for osteoporosis (OP) through a comprehensive synthesis of recent randomized controlled trial (RCT) evidence. Methods A systematic literature search was conducted across PubMed, Web of Science, CNKI, and Wanfang databases (2014 - 2024) to identify RCTs investigating acupuncture combined with conventional therapy for OP. Study quality was appraised using the Cochrane Risk of Bias tool, and meta-analyses were performed using RevMan 5.4 and Stata 15.0, with subgroup analyses stratified by intervention type, population characteristics, and treatment duration. Results 28 RCTs (n=2,758) were included. Meta-analysis revealed acupuncture significantly enhanced bone mineral density (BMD) versus controls: total (SMD = 0.47, p = 0.03), femoral neck (MD = 0.05, p = 0.01), lumbar spine (SMD = 0.40, p < 0.001), Ward's triangle (MD = 0.07, p = 0.02), and hip (SMD = 0.55, p < 0.001), with particularly marked improvements in the postmenopausal osteoporosis subgroup. Acupuncture demonstrated significant improvements in treatment efficacy, biochemical markers, pain scores, and symptom assessments, while reducing adverse events. Warm needle moxibustion outperformed controls in femoral neck (MD = 0.07, p = 0.002) and hip BMD (SMD = 0.87, p < 0.001), while electroacupuncture significantly elevated serum calcium (MD = 0.18, p = 0.02). Short-term interventions (≤ 3 months) demonstrated optimal efficacy. Conclusion Acupuncture demonstrates efficacy and safety as an OP adjuvant therapy. Current evidence is limited by regional bias and methodological heterogeneity. Multicenter, large-sample RCTs are needed to standardize protocols and validate long-term therapeutic efficacy. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42024499354.
Collapse
Affiliation(s)
- Zixin Teng
- Second Clinical Medical College of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Jingwei Zhu
- Second Clinical Medical College of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Kuiwu Li
- Second Clinical Medical College of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Tingting Tong
- Department of Encephalopathy, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Wei Li
- Department of Encephalopathy, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Haoran Chu
- Mingyi Hall, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Peiyang Sun
- Department of Encephalopathy, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| |
Collapse
|
4
|
Liu J, Chen X, Yu X. Unraveling the Role of N6-Methylation Modification: From Bone Biology to Osteoporosis. Int J Med Sci 2025; 22:2545-2559. [PMID: 40520903 PMCID: PMC12163421 DOI: 10.7150/ijms.108763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/25/2025] [Indexed: 06/18/2025] Open
Abstract
N6-methyladenosine (m6A) is the most abundant and reversible epitranscriptomic modification in eukaryotes, playing a pivotal role in regulating various RNA metabolic processes, including splicing, nuclear export, translation and degradation. Emerging evidence indicates that m6A modification is indispensable in biological processes of bone cells such as proliferation, differentiation and apoptosis. Given its pivotal influence on osteoblastogenesis and osteoclastogenesis, m6A modification, particularly via METTL3, has attracted considerable attention in osteoporosis (OP). In this review, we probe the function of m6A modification in intramembranous and endochondral ossification. Furthermore, we summarize the regulatory role of m6A modification in various biological processes in osteoblasts, osteoclasts and osteocytes, focusing on its potential signaling pathways in osteoblast and osteoclast differentiation. Specifically, m6A modulates osteoblast differentiation predominantly through signaling pathways such as Wnt/β-catenin, PI3K/AKT, and BMP/Smad. Concurrently, it regulates osteoclast differentiation and maturation via the RANKL/RANK pathway and its downstream signaling mechanisms. We also discuss recent discoveries that m6A modification regulates OP and further explore its potential clinical value in diagnosing and treating OP. Collectively, m6A modification serves as a crucial regulatory factor in bone metabolism, and a comprehensive understanding of the molecular mechanisms of m6A modification in bone biology is expected to provide new targets for treating OP.
Collapse
Affiliation(s)
| | | | - Xijie Yu
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, No. 37, Guoxue Xiang, Chengdu 610041, China
| |
Collapse
|
5
|
Xiao W, Yike W, Gongwen L, Youjia X. Ferroptosis-mediated immune responses in osteoporosis. J Orthop Translat 2025; 52:116-125. [PMID: 40271049 PMCID: PMC12017889 DOI: 10.1016/j.jot.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Osteoporosis is a common systemic metabolic disease, characterized by decreased bone mass and susceptibility to fragility fractures, often associated with aging, menopause, genetics, and immunity. Ferroptosis plays an underestimated yet crucial role in the further impact of immune function changes on osteoporosis. Cell ferroptosis can induce alterations in immune function, subsequently influencing bone metabolism. In this context, this review summarizes several mechanisms of ferroptosis and introduces the latest insights on how ferroptosis regulates immune responses, exploring the interactions between ferroptosis and other mechanisms such as oxidative stress, inflammation, etc. This review elucidates potential treatment strategies for osteoporosis, emphasizing the promising potential of ferroptosis as an emerging target in the treatment of osteoporosis. In conclusion, preparations related to ferroptosis exhibit substantial clinical promise for enhancing bone mass restoration. The translational potential of this article: This review elucidates a nuanced conversation between the immune system and osteoporosis, with ferroptosis serving as the connecting link. These findings underscore the potential of ferroptosis inhibition as a therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Wang Xiao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wang Yike
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liu Gongwen
- Department of Orthopaedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xu Youjia
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
6
|
Jin Y, Wang Y, Wang C, Zhang L, Gao D, Liu H, Cao Q, Tian C, Bian Y, Wang Y. Salidroside inhibits osteoclast differentiation based on osteoblast-osteoclast interaction via HIF-1a pathway. Chin J Nat Med 2025; 23:572-584. [PMID: 40383613 DOI: 10.1016/s1875-5364(25)60864-8] [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: 09/21/2024] [Revised: 10/25/2024] [Accepted: 01/02/2025] [Indexed: 05/20/2025]
Abstract
This study investigated the regulatory potential of salidroside (SAL), a primary active compound in Rhodiola rosea L., on osteoclast differentiation by modulating the hypoxia-inducible factor 1-alpha (HIF-1a) pathway in osteoblasts. Luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were employed to validate whether the receptor activator of nuclear factor-?B ligand (RANKL) is the downstream target gene of HIF-1a in osteoblasts. The study also utilized lipopolysaccharide (LPS)-induced mouse osteolysis to examine the impact of SAL on osteolysis in vivo. Furthermore, conditioned medium (CM) from SAL-pretreated osteoblasts was used to investigate the paracrine effects on osteoclastogenesis through the HIF-1a pathway. Hypoxic condition-induced overexpression of HIF-1a upregulated RANKL levels by binding to the RANKL promoter and enhancing transcription in osteoblastic cells. In vivo, SAL significantly alleviated bone tissue hypoxia and decreased the expression of HIF-1a by downregulating the expression of RANKL, vascular endothelial growth factor (VEGF), interleukin 6 (IL-6), and angiopoietin-like 4 (ANGPTL4). In the paracrine experiment, conditioned media from SAL-pretreated osteoblasts inhibited differentiation through the HIF-1a/RANKL, VEGF, IL-6, and ANGPTL4 pathways. RANKL emerges as the downstream target gene regulated by HIF-1a in osteoblasts. SAL significantly alleviates bone tissue hypoxia and bone loss in LPS-induced osteolysis through the HIF-1a/RANKL, VEGF, IL-6, and ANGPTL4 pathways. SAL inhibits osteoclast differentiation by regulating osteoblast paracrine secretion.
Collapse
Affiliation(s)
- Yutong Jin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yao Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chuan Wang
- Department of Stomatology, NHC Key Laboratory of Hormones and Development, Chu Hsien- I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Department of Stomatology, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300134, China
| | - Lingling Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Dandan Gao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Haizhao Liu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qingwen Cao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chenchen Tian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Yue Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
7
|
Luo P, Zhong Y, Yang X, Lai Q, Huang S, Zhang X, Zhang B, Wei Y. Self-assembled water soluble and bone-targeting phosphorylated quercetin ameliorates postmenopausal osteoporosis in ovariectomy mice. Colloids Surf B Biointerfaces 2025; 249:114495. [PMID: 39798316 DOI: 10.1016/j.colsurfb.2025.114495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
Natural compounds have shown promising application prospects in preventing or treating various diseases, including osteoporosis on account of their abundant sources, low price, multi-targeting and multiple biological effects. As a bioactive natural product, quercetin (Que) has previously demonstrated to ameliorate osteoporosis (OP), however, its poor bioavailability resulting from low water solubility, poor stability and lack of bone-targeting largely restricted its efficacy and clinical applications. Inspired by the bone-targeting capability of phosphate compounds, we reported a one-step procedure for synthesis of phosphorylated Que (p-Que) by direct phosphorylating phenol groups of Que for the first time. The phosphate groups on p-Que could not only improve the water dispersibility of Que, but also endow p-Que desirable bioavailability and bone-targeting feature. The results from biological assays suggested that p-Que could inhibit osteoclastogenesis and bone resorption and alleviate trabeculae loss in osteoporotic mice. In conclusion, this work demonstrated that phosphorylation strategy can effectively solve low water solubility, lack of bone-targeting capability and poor bioavailability of natural compounds, providing a novel and efficient approach for development of OP nanomedicines.
Collapse
Affiliation(s)
- Peng Luo
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China; Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Yanlong Zhong
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China; Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Xiaowei Yang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China; Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Qi Lai
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China; Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Shaorong Huang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, China.
| | - Xiaoyong Zhang
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China.
| | - Bin Zhang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China.
| | - Yen Wei
- Department of Chemistry and the Tsinghua Center for Frontier Polymer Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
8
|
Cheng H, Chen H, Yan X, Zhang Q. Inhibition of NEURL3 Suppresses Osteoclast Differentiation via BMP7 Ubiquitination Modulation. Appl Biochem Biotechnol 2025; 197:3466-3481. [PMID: 39960614 DOI: 10.1007/s12010-025-05198-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 05/11/2025]
Abstract
Osteoporosis (OP) is a genetic disorder characterized by an imbalance between osteoblast-mediated bone formation and osteoclast-induced bone resorption. However, the underlying gene-related mechanisms of its pathogenesis remain to be fully elucidated. Aberrantly expressed neuralized E3 ubiquitin-protein ligase 3 (NEURL3), which is related to osteoclastic differentiation, was identified through the analysis of the microarray profile GSE176265. Bone marrow-derived macrophages (BMMs) were isolated from the femurs and tibias of C57BL/6 J mice and treated with 30 ng/mL macrophage-colony-stimulating factor (M-CSF) and 100 ng/mL receptor activator of nuclear factor-kappa B ligand (RANKL) to induce osteoclastic differentiation, thereby mimicking OP in vitro. To model OP in vivo, ovariectomy (OVX)-induced bone loss was performed in mice. High expression levels of NEURL3 were confirmed in clinical samples, OP model cells, and OP model mice using quantitative real-time polymerase chain reaction (qRT-PCR). The impact of NEURL3 on osteoclastic differentiation was assessed by evaluating cell viability and the expression levels of osteoclastogenesis-related marker genes. Additionally, bone loss in mice was quantified using micro-computed tomography before and after NEURL3 inhibition. Mechanistically, the effects of NEURL3 on osteogenic differentiation were investigated by determining the protein levels of osteogenic markers via Western blotting. NEURL3 was markedly overexpressed in serum samples collected from patients with OP, OVX-induced OP mouse models, and induced osteoclasts. Inhibition of NEURL3 leads to a 20% decrease in BMM survival rate and a reduction in the number of tartrate-resistant acid phosphatase (TRAP) positive cells, which is a characteristic of mature osteoclasts. Furthermore, the expression levels of osteoclastogenesis-related marker genes were reduced by 50%. In vivo studies revealed that suppressing NEURL3 resulted in a 38% improvement in trabecular bone volume (BV/TV) and a 28% increase in bone mineral density (BMD) in the OVX-induced OP mice. Mechanistically, NEURL3 promoted osteoclast differentiation by increasing the ubiquitination levels of BMP7. Inhibition of BMP7 reversed the effects of NEURL3 on osteoclast differentiation in BMMs. Suppression of NEURL3 inhibits osteoclast differentiation of BMMs in vitro and alleviates bone loss in vivo. The underlying mechanism may involve NEURL3-induced ubiquitination of BMP7. Collectively, the downregulation of NEURL3 represents a promising therapeutic strategy for suppressing osteoclast differentiation and treating OP.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Spine Surgery, Jinhua Municipal Central Hospital, No.365, East Renmin Road, Wucheng District, Jinhua, 321000, Zhejiang, China.
| | - Huilan Chen
- Department of Geriatrics, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Xin Yan
- Department of Geriatrics, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Qizhe Zhang
- Department of Geriatrics, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| |
Collapse
|
9
|
Tang C, Lv X, Zou L, Rong Y, Zhang L, Xu M, Li S, Chen G. Cadmium exposure and osteoporosis: epidemiological evidence and mechanisms. Toxicol Sci 2025; 205:1-10. [PMID: 40127184 DOI: 10.1093/toxsci/kfaf031] [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] [Indexed: 03/26/2025] Open
Abstract
Cadmium (Cd) is a toxic heavy metal with a long biological half-life, exerting adverse effects on most tissues and organs in the human body. Inhalation, ingestion, and skin contact are the main ways of exposure to Cd. Bone is one of the target organs of Cd. The aging of the population has been considered as the reason for the high incidence rate of osteoporosis, but recent studies have emphasized that the risk of osteoporosis is related to Cd exposure. With the widespread use of Cd-containing materials in industrial and agricultural activities, the risk of Cd exposure is worrying. This review covers the epidemiological, in vivo, and in vitro studies on Cd exposure and osteoporosis. Epidemiological evidence has emphasized a positive association between Cd exposure and the occurrence rates of osteoporosis and fractures. Experimental studies have demonstrated that Cd induces osteoporosis through both direct and indirect pathways. The indirect pathway encompasses inducing renal dysfunction to impair calcium and phosphorus metabolism, whereas the direct pathway consists of directly influencing bone cells. This review aims to emphasize that Cd exposure may be an overlooked risk factor for osteoporosis and to elucidate the direct and indirect molecular mechanisms by which Cd induces osteoporosis. Understanding the pathogenesis of Cd-induced osteoporosis is crucial for the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Cai Tang
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Xingmin Lv
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Lingling Zou
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yi Rong
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Lu Zhang
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Maoting Xu
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Sheng Li
- Southwest Medical University, Luzhou 646000, China
| | - Guiquan Chen
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
10
|
Wang H, Yuan X, Han J, Wu Z, Ma Z, Shi F, Luo Z, Chen Z, Guo C, Yuan G, He X, Ling Z, Meng L, Shen R, Huang J, Xu R. RO5126766 attenuates osteoarthritis by inhibiting osteoclastogenesis and protecting chondrocytes through mediating the ERK pathway. J Orthop Translat 2025; 52:27-39. [PMID: 40231159 PMCID: PMC11995706 DOI: 10.1016/j.jot.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/09/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
Background Osteoarthritis (OA) is a degenerative joint disease that remains challenging to treat due to lack of complete understanding of its pathogenesis. Previous studies have identified RO5126766 (RO) as a small molecule compound that inhibited RAF/MEK-ERK pathway and garnered much interest for its anti-cancer properties. But its role in the treatment of OA remains unclear. Methods This study employed the anterior cruciate ligament transection (ACLT) procedure to create an OA model in mice. The effects of RO on pathological changes in articular cartilage and subchondral bone were assessed using micro-CT and histological staining. Mice received peritoneal injections of RO at 1 mg/kg and 5 mg/kg biweekly for 4 weeks after ACLT, while control mice received saline. In vitro, bone marrow-derived macrophages were cultured to examine the effects of RO on osteoclast activation using immunofluorescence, TRAP staining, and bone resorption assays. The inflammatory degeneration of chondrocytes and gene expression levels were evaluated using staining and RT-qPCR. Western blot and immunohistochemistry were used to analyze MAPK signaling and autophagy-related protein expression, investigating RO's molecular mechanism in OA treatment. Human single-cell data were also analyzed to identify genes and pathways upregulated in OA tissues. Results Our findings showed that RO protects subchondral bone by inhibiting osteoclast formation in the ACLT mouse model of OA. In vitro, RO was shown to inhibit osteoclast differentiation and reduce inflammatory degeneration of chondrocytes. Mechanistically, RO counteracted subchondral osteoclast hyperactivation by suppressing the ERK/c-fos/NFATc1 signaling pathway. Additionally, RO inhibited LPS-induced inflammatory degeneration of chondrocytes and enhanced autophagy via the ERK pathway. Single-cell analysis further confirmed significant upregulation of the ERK signaling pathway in human OA tissues. Conclusions Overall, our findings suggested that RO inhibited osteoclast differentiation and protected articular cartilage, suggesting its potential as a novel treatment for OA. Translational potential of this article In this study, we have, for the first time, substantiated the therapeutic potential of RO in the treatment of OA. By demonstrating its ability to inhibit osteoclast differentiation and protect articular cartilage, RO could offer a new avenue for disease-modifying treatments in OA. Thus, this paper provides valuable insights into understanding the molecular mechanisms and treatment of OA.
Collapse
Affiliation(s)
- Han Wang
- Department of Orthopedics, Chenggong Hospital of Xiamen University (the 73rd Group Military Hospital of People's Liberation Army), School of Medicine, Xiamen University, Xiamen, 361003, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xiwen Yuan
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Jie Han
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Zuoxing Wu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Zheru Ma
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Fan Shi
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Zhengqiong Luo
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Zihan Chen
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Chenyang Guo
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Guixin Yuan
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xuemei He
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Zemin Ling
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lin Meng
- Department of Electronic and Computer Engineering, Ritsumeikan University, Shiga, 525-8577, Japan
| | - Rong Shen
- Department of Orthopedics, Chenggong Hospital of Xiamen University (the 73rd Group Military Hospital of People's Liberation Army), School of Medicine, Xiamen University, Xiamen, 361003, China
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Jianming Huang
- Department of Orthopedics, Chenggong Hospital of Xiamen University (the 73rd Group Military Hospital of People's Liberation Army), School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Ren Xu
- Department of Orthopedics, Chenggong Hospital of Xiamen University (the 73rd Group Military Hospital of People's Liberation Army), School of Medicine, Xiamen University, Xiamen, 361003, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361000, China
| |
Collapse
|
11
|
Liang H, Yin G, Feng D, Shi G, Chen H, Liu X, Li J. Ras-proximate-1 (RAP1): a prognosis and therapeutic target in the metastatic spread of breast cancer. Clin Exp Metastasis 2025; 42:23. [PMID: 40214810 DOI: 10.1007/s10585-025-10342-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 03/24/2025] [Indexed: 06/11/2025]
Abstract
Breast cancer (BC), a highly heterogeneous disease, has demonstrated a gradual increase in both incidence and mortality rates. At present, it has become one of the most common malignant tumors and the main cause of cancer death worldwide. While early screening is recognized as an effective preventive and therapeutic measure for BC, the disease continues to exhibit a high rate of metastasis. Metastatic BC is still the main cause of poor prognosis and death of patients, necessitating urgent investigation and resolution. Among the various metastatic sites of BC, bone metastases warrant particular attention due to their prevalence. In numerous studies on BC bone metastasis mechanisms, cancer markers have been shown to significantly influence the pattern and extent of BC metastasis and dissemination. In the tumor microenvironment, Ras-proximate-1 (RAP1), a GTPase protein, is not only upregulated in various malignant tumors and bone-related diseases, including BC, but also regulates migration, invasion, distant metastasis, and other signaling pathways in numerous malignant tumor cells, including BC as well. Despite these findings, there remains a paucity of advanced research and discussion on the relationship between RAP1 and BC bone metastasis. Furthermore, no clinically approved RAP1-related inhibitors for BC bone metastasis are currently available. Nevertheless, RAP1 and its associated signaling molecules represent potential molecular targets for the prevention and treatment of BC bone metastasis, warranting further investigation. Therefore, this article provides a comprehensive review of RAP1's pathogenic role in BC bone metastasis, emphasizes RAP1 and its associated signaling pathways, and summarizes current research on natural compounds and extracts that modulate BC bone metastasis via RAP1 or RAP1-related signaling pathways. This review aims to offer novel perspectives for developing RAP1 as a potential molecular target in the prevention and treatment of BC bone metastasis, as well as for the development of related therapeutic agents.
Collapse
Affiliation(s)
- Hongyi Liang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Dandan Feng
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Guangxi Shi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, Shandong, 250014, China
| | - Hanhan Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, Shandong, 250014, China
| | - Xiaofei Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, Shandong, 250014, China
| | - Jingwei Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, Shandong, 250014, China.
| |
Collapse
|
12
|
Lin Y, Jiang S, Yao Y, Li H, Jin H, Yang G, Ji B, Li Y. Posttranslational Modification in Bone Homeostasis and Osteoporosis. MedComm (Beijing) 2025; 6:e70159. [PMID: 40170748 PMCID: PMC11959162 DOI: 10.1002/mco2.70159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025] Open
Abstract
Bone is responsible for providing mechanical protection, attachment sites for muscles, hematopoiesis micssroenvironment, and maintaining balance between calcium and phosphorate. As a highly active and dynamically regulated organ, the balance between formation and resorption of bone is crucial in bone development, damaged bone repair, and mineral homeostasis, while dysregulation in bone remodeling impairs bone structure and strength, leading to deficiency in bone function and skeletal disorder, such as osteoporosis. Osteoporosis refers to compromised bone mass and higher susceptibility of fracture, resulting from several risk factors deteriorating the balanced system between osteoblast-mediated bone formation and osteoclast-mediated bone resorption. This balanced system is strictly regulated by translational modification, such as phosphorylation, methylation, acetylation, ubiquitination, sumoylation, glycosylation, ADP-ribosylation, S-palmitoylation, citrullination, and so on. This review specifically describes the updating researches concerning bone formation and bone resorption mediated by posttranslational modification. We highlight dysregulated posttranslational modification in osteoblast and osteoclast differentiation. We also emphasize involvement of posttranslational modification in osteoporosis development, so as to elucidate the underlying molecular basis of osteoporosis. Then, we point out translational potential of PTMs as therapeutic targets. This review will deepen our understanding between posttranslational modification and osteoporosis, and identify novel targets for clinical treatment and identify future directions.
Collapse
Affiliation(s)
- Yuzhe Lin
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaChina
- Xiangya School of Medicine Central South UniversityChangshaChina
| | - Shide Jiang
- The Central Hospital of YongzhouYongzhouChina
| | - Yuming Yao
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Hengzhen Li
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Hongfu Jin
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Guang Yang
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Bingzhou Ji
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Yusheng Li
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
13
|
Chen J, Liao Y, Sheng Y, Yao H, Li T, He Z, Ye WWY, Yin M, Tang H, Zhao Y, Zhang P, Wang Y, Fu X, Ji Y. FSH exacerbates bone loss by promoting osteoclast energy metabolism through the CREB-MDH2-NAD + axis. Metabolism 2025; 165:156147. [PMID: 39880362 DOI: 10.1016/j.metabol.2025.156147] [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: 11/21/2024] [Revised: 01/07/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
AIMS Osteoclast energy metabolism is a promising target for treating diseases characterized by high osteoclast activity, such as osteoporosis. However, the regulatory factors involved in osteoclast bioenergetic processes are still in the early stages of being fully understood. This study reveals the effects of follicle-stimulating hormone (FSH) on osteoclast energy metabolism. METHODS The Lyz2-Cre-Flox model selectively deletes FSH receptor (FSHR) from osteoclast precursor cells to generate Fshrf/f; Lyz2-Cre (Fshrf/f; Cre) mice. Bone quality was assessed using micro-computed tomography, histomorphometric analysis, and dual-fluorescence labeling. The in vitro assays measured oxygen consumption rate, extracellular acidification rate, pyruvate content, and mitochondrial membrane potential to determine metabolic flux. RNA-seq, LC-MS, dual-luciferase reporter assays, and chromatin immunoprecipitation (ChIP) assays were used to elucidate the underlying mechanisms. RESULTS FSHR deficiency in osteoclasts protected bone from resorption under normal and ovariectomized conditions. FSHR-deficient osteoclasts have reduced nicotinamide adenine dinucleotide (NAD+) levels, impairing osteoclast activity and energy metabolism. Mechanistically, FSH influenced NAD+ levels via the CREB/MDH2 axis. Treatment with FSH monoclonal antibodies rescued bone loss in OVX mice and reduced bone marrow NAD+ levels. CONCLUSIONS Targeting FSH may be a promising metabolic modulation strategy for treating osteoporosis and other diseases associated with high osteoclast activity.
Collapse
Affiliation(s)
- Jingqiu Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Yilin Liao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Yue Sheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Hantao Yao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Ting Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Zhenru He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Weng Wan Yue Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Mengjie Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Huilin Tang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Yaoyu Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Peiqi Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Yuting Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Xiazhou Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China.
| | - Yaoting Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China.
| |
Collapse
|
14
|
Mamet T, Yang J, Zhang J, Guo Y, Zhao Z. Yak milk inhibits osteoclast differentiation by suppressing TRPV5 expression. J Dairy Sci 2025; 108:3142-3150. [PMID: 39824495 DOI: 10.3168/jds.2024-25607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/17/2024] [Indexed: 01/20/2025]
Abstract
Yak milk is a potential nutrient for improving osteoporosis. However, the effect of yak milk on the expression of Ca2+ion channel TRPV5 during osteoclast differentiation is still unclear. This study used ruthenium red as a control to investigate the effect of yak milk on osteoclast differentiation and activity. Tartrate-resistant acid phosphatase staining and bone resorption pit experiments showed that yak milk inhibited osteoclast differentiation and bone resorption activity in a dose-dependent manner. In addition, yak milk can inhibit osteoclast activity by inhibiting the expression of TRPV5. Quantitative real-time PCR and western blot results also exhibited that yak milk significantly decreased the expression of TRPV5 and calbindin-D28k mRNA and protein in osteoclasts. These results suggest that yak milk inhibits nuclear factor-κβ ligand-receptor activator-induced osteoclast differentiation and bone resorption activity in RAW 264.7 cells by suppressing the expression level of TRPV5 and calbindin-D28k mRNA and protein.
Collapse
Affiliation(s)
- Torkun Mamet
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830046, China.
| | - Jingru Yang
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Jin Zhang
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Yanping Guo
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Zhongkai Zhao
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830046, China
| |
Collapse
|
15
|
Guo ZY, Yin NN, Li XF, Wang MM, Sui XN, Jiang CD, Xu MH, Jia XE, Fu CJ, Chen TL, Liu X. Exosomes secreted from M2-polarized macrophages inhibit osteoclast differentiation via CYLD. Tissue Cell 2025; 93:102645. [PMID: 39671756 DOI: 10.1016/j.tice.2024.102645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 10/31/2024] [Accepted: 11/27/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE Bone resorption mediated by osteoclast differentiation induces the occurrence of bone-related diseases. Macrophages, an origin of osteoclasts, whose M2 type can reduce inflammation-induced bone damage. We aimed to investigate the effect of M2 macrophage-derived exosomes on osteoclast formation and elucidate its underlying mechanism. MATERIALS AND METHODS Exosomes were isolated from M2 macrophages (M2-exo) and were used to treat osteoclast-like cells. Osteoclast formation was evaluated using tartrate-resistant acid phosphatase, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blotting. The molecular mechanism of M2-exo function was analyzed by qRT-PCR, phosphor-kinase array analysis, and Western blotting. RESULTS M2-exo was internalized by osteoclasts and inhibited osteoclast differentiation in vitro. Moreover, CYLD was highly expressed in M2 macrophages and M2-exo-treated osteoclasts, and knockdown of it abrogated the inhibition of osteoclast differentiation caused by M2-exo. Additionally, CYLD suppressed the phosphorylation of STAT3, and STAT3 activator colivelin reversed the inhibition of osteoclast differentiation induced by CYLD overexpression. CONCLUSION M2-exo inhibits osteoclast differentiation via delivering CYLD, which inactivates STAT3 signaling. These findings may provide a novel therapeutic option for bone diseases including periodontitis.
Collapse
Affiliation(s)
- Zi-Yan Guo
- Department of Stomatology, Changhai Hospital Affiliated to Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Nan-Nan Yin
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, China
| | - Xiao-Fei Li
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, China
| | - Meng-Meng Wang
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, China
| | - Xiao-Na Sui
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, China
| | - Cai-di Jiang
- Department of Stomatology, Changhai Hospital Affiliated to Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Ming-Hua Xu
- Department of Stomatology, Changhai Hospital Affiliated to Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xiao-E Jia
- Department of Stomatology, Changhai Hospital Affiliated to Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Chong-Jian Fu
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, China.
| | - Tie-Lou Chen
- Department of Periodontal, Military Dental Center, Changhai Hospital Affiliated to Naval Medical University, 168 Changhai Road, Yang Pu District, Shanghai, 200433, China.
| | - Xin Liu
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, China.
| |
Collapse
|
16
|
Wen G, Li H, Yang J, Mai B, Zhou T, Mo G, Li Y, Lai Y. Isonardosinone attenuates osteoclastogenesis and OVX-induced bone loss via the MAPK/NF-κB pathway. Toxicol Appl Pharmacol 2025; 497:117267. [PMID: 39956462 DOI: 10.1016/j.taap.2025.117267] [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: 12/17/2024] [Revised: 02/03/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Osteoporosis is a globally prevalent metabolic bone disease that manifests itself as a decrease in bone mineral density and deterioration of bone structure, which reduces overall bone strength and increases fracture risk. However, the effect of anti-inflammatory isonardosinone (the active ingredient in Nardostachys chinensis) on osteoclastogenesis is unknown. We first predicted the main pathways and targets of ISO action in osteoporosis by network pharmacology. CCK-8 was used to test whether ISO affects cell proliferation of BMMs (osteoclast precursor cells) and to determine the safe action concentration. TRAcP and F-actin staining were used to characterise the inhibitory effect on osteoclast differentiation. RT-PCR and WB were used to examine changes in the relative expression of genes and proteins generated by osteoclasts under isopinacolone treatment, and we examined its effects on the RANKL-activated MAPK and NF-κB signaling pathways. An ovariectomy-induced osteoporosis model was constructed to assess the in vivo therapeutic effects of ISO. CCK-8 results showed that ISO had no cytotoxic or proliferative effects on BMMs at concentrations below 30 μM; TRAcP staining showed that ISO suppressed osteoclastogenesis in a concentration- and time-gradient-dependent manner; and F-actin staining showed that ISO suppressed osteoblast skeleton formation and expansion; RT-PCR and Western Blot assays showed that ISO suppressed the expression of CTSK, NFATC1, MMP9, C-Fos, and ACP5, inhibited the phosphorylation of JNK, P38, and ERK, and reversed the degradation of IκB-α, especially within 15 min. The in vivo results indicated that ISO has therapeutic effects on osteoporosis by improving bone microstructure to rescue bone loss. Taken together, these results lead to the conclusion that ISO is an attractive drug development strategy for the treatment of osteoporosis by effectively suppressing osteoclastogenesis through the MAPK/NF-κB signaling pathway, thereby reversing the bone loss associated with ovariectomy in vivo.
Collapse
Affiliation(s)
- Guangwei Wen
- Guangzhou Panyu District Hualong Hospital, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China; Liwan District Orthopedics Hospital of Guangzhou, Spinal Orthopedics, Guangdong, China.
| | - Haishan Li
- Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Jiasheng Yang
- Guangzhou University of Chinese Medicine, Guangzhou, China; Liwan District Orthopedics Hospital of Guangzhou, Spinal Orthopedics, Guangdong, China
| | - Bin Mai
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tengpeng Zhou
- Guangzhou University of Chinese Medicine, Guangzhou, China; Liwan District Orthopedics Hospital of Guangzhou, Spinal Orthopedics, Guangdong, China
| | - GuoYe Mo
- Guangzhou University of Chinese Medicine, Guangzhou, China; The First Affiliated Hospital of Guangzhou University of Chinese medicine, Spinal Orthopedics, Guangzhou, China
| | - Yongxian Li
- Guangzhou University of Chinese Medicine, Guangzhou, China; The First Affiliated Hospital of Guangzhou University of Chinese medicine, Spinal Orthopedics, Guangzhou, China
| | - Yiyi Lai
- Guangzhou University of Chinese Medicine, Guangzhou, China; Liwan District Orthopedics Hospital of Guangzhou, Spinal Orthopedics, Guangdong, China.
| |
Collapse
|
17
|
Lu J, He Q, Wang H, Yao L, Duffy M, Guo H, Braun C, Zhou Y, Liang Q, Lin Y, Bandyopadhyay S, Tan K, Choi Y, Liu XS, Qin L. Bone marrow adipogenic lineage precursors are the major regulator of bone resorption in adult mice. Bone Res 2025; 13:39. [PMID: 40102423 PMCID: PMC11920254 DOI: 10.1038/s41413-025-00405-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/28/2024] [Accepted: 01/20/2025] [Indexed: 03/20/2025] Open
Abstract
Bone resorption by osteoclasts is a critical step in bone remodeling, a process important for maintaining bone homeostasis and repairing injured bone. We previously identified a bone marrow mesenchymal subpopulation, marrow adipogenic lineage precursors (MALPs), and showed that its production of RANKL stimulates bone resorption in young mice using Adipoq-Cre. To exclude developmental defects and to investigate the role of MALPs-derived RANKL in adult bone, we generated inducible reporter mice (Adipoq-CreER Tomato) and RANKL deficient mice (Adipoq-CreER RANKLflox/flox, iCKO). Single cell-RNA sequencing data analysis and lineage tracing revealed that Adipoq+ cells contain not only MALPs but also some mesenchymal progenitors capable of osteogenic differentiation. In situ hybridization showed that RANKL mRNA is only detected in MALPs, but not in osteogenic cells. RANKL deficiency in MALPs induced at 3 months of age rapidly increased trabecular bone mass in long bones as well as vertebrae due to diminished bone resorption but had no effect on the cortical bone. Ovariectomy (OVX) induced trabecular bone loss at both sites. RANKL depletion either before OVX or at 6 weeks post OVX protected and restored trabecular bone mass. Furthermore, bone healing after drill-hole injury was delayed in iCKO mice. Together, our findings demonstrate that MALPs play a dominant role in controlling trabecular bone resorption and that RANKL from MALPs is essential for trabecular bone turnover in adult bone homeostasis, postmenopausal bone loss, and injury repair.
Collapse
Affiliation(s)
- Jiawei Lu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qi He
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Huan Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael Duffy
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hanli Guo
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Corben Braun
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yilu Zhou
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qiushi Liang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yuewei Lin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shovik Bandyopadhyay
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Kai Tan
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Yongwen Choi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - X Sherry Liu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
18
|
Sun H, Liu Y, Huang Y, Xiong K, Zhang Z, Wang W, Dai Y, Li J, Li Q, Wang S, Shi C. Echinococcus granulosus sensu lato promotes osteoclast differentiation through DUSP4-MAPK signaling in osseous echinococcosis. Front Microbiol 2025; 16:1558603. [PMID: 40177487 PMCID: PMC11961949 DOI: 10.3389/fmicb.2025.1558603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Osseous echinococcosis, caused by Echinococcus granulosus infection, is characterized by progressive bone destruction driven by abnormal osteoclast activation. Dual-specificity phosphatase 4 (DUSP4), a key negative regulator of the MAPK pathway, inhibits osteoclast differentiation and bone resorption. This study aimed to elucidate the role of DUSP4 in E. granulosus-induced bone loss. Methods In vitro, a co-culture system of E. granulosus protoscoleces (PSCs) and bone marrow-derived macrophages (BMMs) was established. Osteoclast differentiation and bone resorption were assessed using TRAP staining and F-actin immunofluorescence. Transcriptome sequencing identified DUSP4 as a key regulator. DUSP4 overexpression was performed to evaluate its effects on osteoclast markers and MAPK signaling (ERK, JNK, p38). In vivo, a mouse model of osseous echinococcosis was developed, and DUSP4 overexpression was achieved via lentiviral transduction. Bone destruction was analyzed using X-ray, micro-CT, and histology. Results PSCs significantly enhanced osteoclast differentiation and bone resorption, upregulated osteoclast markers (CTSK, NFATc1), and activated MAPK signaling. DUSP4 overexpression reversed these effects, reducing osteoclast activity and MAPK phosphorylation. In vivo, PSC infection caused severe bone destruction, which was mitigated by DUSP4 overexpression. Disscussion This study reveals the molecular mechanism by which Echinococcus granulosus drives abnormal osteoclast activation through the DUSP4-MAPK signaling axis. Parasitic infection suppresses DUSP4 expression, relieving its negative regulation of the MAPK pathway and leading to excessive osteoclast differentiation. Restoring DUSP4 expression effectively reverses abnormal MAPK pathway activation, reducing osteoclast bone resorption activity to physiological levels. These findings not only provide new insights into the pathological mechanisms of bone destruction in osseous echinococcosis but also establish DUSP4 as a critical therapeutic target for pathological bone resorption, laying the groundwork for host-directed treatment strategies for parasitic bone diseases.
Collapse
Affiliation(s)
- Haohao Sun
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Yaqing Liu
- The First Affiliated Hospital of Shihezi University, Shihezi, China
- The Medical College of Shihezi University, Shihezi, China
| | - Yiping Huang
- The Medical College of Shihezi University, Shihezi, China
| | - Kangjun Xiong
- The Medical College of Shihezi University, Shihezi, China
| | - Zhendong Zhang
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Weishan Wang
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Yi Dai
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Jing Li
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Qi Li
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Sibo Wang
- Xi’an Jiaotong University Affiliated Honghui Hospital, Xi’an, China
| | - Chenhui Shi
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| |
Collapse
|
19
|
Qiu D, Yan B, Xue H, Xu Z, Tan G, Liu Y. Perspectives of exosomal ncRNAs in the treatment of bone metabolic diseases: Focusing on osteoporosis, osteoarthritis, and rheumatoid arthritis. Exp Cell Res 2025; 446:114457. [PMID: 39986599 DOI: 10.1016/j.yexcr.2025.114457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/13/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Bone metabolic disorders, constituting a group of prevalent and grave conditions, currently have a scarcity of therapeutic alternatives. Over the recent past, exosomes have been at the forefront of research interest, owing to their nanoparticulate nature and potential for therapeutic intervention. ncRNAs are a class of heterogeneous transcripts that they lack protein-encoding capacity, yet they can modulate the expression of other genes through multiple mechanisms. Mounting evidence underscores the intricate role of exosomes as ncRNAs couriers implicated in the pathogenesis of bone metabolic disorders. In this review, we endeavor to elucidate recent insights into the roles of three ncRNAs - miRNAs, lncRNAs, and circRNAs - in bone metabolic ailments such as osteoporosis, osteoarthritis, and rheumatoid arthritis. Additionally, we examine the viability of exosomal ncRNAs as innovative, cell-free modalities in the diagnosis and therapeutic management of bone metabolic disorders. We aim to uncover the critical function of exosomal ncRNAs within the context of bone metabolic diseases.
Collapse
Affiliation(s)
- Daodi Qiu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Binghan Yan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Haipeng Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhanwang Xu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Guoqing Tan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yajuan Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250300, China.
| |
Collapse
|
20
|
Li N, Mu X, Zhang S, Wang H. Recent advances in the multifaceted mechanisms of catalpol in treating osteoporosis. Front Pharmacol 2025; 16:1560715. [PMID: 40103589 PMCID: PMC11913683 DOI: 10.3389/fphar.2025.1560715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
Catalpol (CAT) is a landmark active ingredient in traditional Chinese medicine Rehmannia (TCT), also known as dehydroxybenzoate catalpone, which is a kind of iridoid terpene glycoside with strong antioxidant, anti-inflammatory, antitumor and other biological activities. It can exert its anti-disease effect in a variety of ways. For some patients with chronic diseases, the application of azalea alcohol in rehmannia may bring more comprehensive and long-lasting efficacy. Studies have shown that the anti-disease effect of catalpol in osteoporosis (OP) is mainly achieved through various pathways such as Wnt/β-catenin signaling pathways to promote osteogenic differentiation, and RANKL/RANK and other signaling pathways to inhibit osteoclastic differentiation. At present, there is a slight lack of analysis of the mechanism of action of catalpa alcohol in the treatment of osteoporosis, so this study comprehensively searched the literature on the mechanism of action of catalpa alcohol in the treatment of osteoporosis in various databases, and reviewed the research progress of its role and mechanism, to provide reference and theoretical basis for the further development and application of catalpol.
Collapse
Affiliation(s)
- Na Li
- Shandong Co-Innovation Center of Classic Traditional Chinese Medicine Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoying Mu
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | | | - Huaxin Wang
- Shandong Co-Innovation Center of Classic Traditional Chinese Medicine Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
21
|
Tchouto MN, Bucher CH, Mess AK, Haas S, Schmidt-Bleek K, Duda GN, Beule D, Milek M. Pronounced impairment of B cell differentiation during bone regeneration in adult immune experienced mice. Front Immunol 2025; 16:1511902. [PMID: 40098964 PMCID: PMC11911212 DOI: 10.3389/fimmu.2025.1511902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Alterations of the adaptive immune system have been shown to impact bone healing and may result in impaired healing in some patients. Apart from T cells, B cells are the key drivers of adaptive immunity. Therefore, their role in age-associated impairments of bone healing might be essential to understand delays during the healing process. B cells are essential for bone formation, and their dysfunction has been associated with aging or autoimmune diseases. But whether age-associated changes in B cell phenotypes are involved in bone regeneration is unknown. Methods Here, we aimed to characterize the role of immune aging in B cell phenotypes during the early inflammatory phase of bone healing. By comparing non-immune experienced with young and immune experienced mice we aimed to analyze the effect of gained immune experience on B cells. Our single cell proteo-genomics analysis quantified thousands of transcriptomes of cells that were isolated from post osteotomy hematoma and the proximal and distal bone marrow cavities, and enabled us to evaluate cell proportion, differential gene expression and cell trajectories. Results While the B cell proportion in young and non-immune experienced animals did not significantly change from 2 to 5 days post osteotomy in the hematoma, we found a significant decrease of the B cell proportion in the immune experienced mice, which was accompanied by the decreased expression of B cell specific genes, suggesting a specific response in immune experienced animals. Furthermore, we detected the most extensive B cell differentiation block in immune-experienced mice compared to non-immune experienced and young animals, predominantly in the transition from immature to mature B cells. Discussion Our results suggest that the pronounced impairment of B cell production found in immune experienced animals plays an important role in the initial phase leading to delayed bone healing. Therefore, novel therapeutic approaches may be able target the B cell differentiation defect to retain B cell functionality even in the immune experienced setting, which is prone to delayed healing.
Collapse
Affiliation(s)
- Mireille Ngokingha Tchouto
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian H. Bucher
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ann-Kathrin Mess
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Simon Haas
- Systems Hematology, Stem Cells & Precision Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Miha Milek
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
22
|
Gajewski D, Hennig AF, Grün R, Siggelkow H, Vishnolia S, Bastian L, Taipaleenmäki H, Schulz A, Kornak U, Hesse E. Paradoxical combination of osteosclerosis and osteopenia in an adult woman with biallelic TNFRSF11A loss-of-function variants escaping nonsense-mediated decay. JBMR Plus 2025; 9:ziae179. [PMID: 39906258 PMCID: PMC11788561 DOI: 10.1093/jbmrpl/ziae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/29/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Osteoclasts are essential for bone resorption, playing a crucial role in skeletal development, homeostasis, and remodeling. Their differentiation depends on the RANK receptor encoded by the TNFRSF11A gene, with defects in this gene linked to osteoclast-poor sclerosing skeletal dysplasias. This report presents a 37-yr-old woman with normal height, valgus deformities that were treated surgically, frequent fractures, scoliosis, mildly elevated BMD, sclerotic diaphyseal bone, and metaphyseal widening. Initially suspected of having dysosteosclerosis, her diagnosis shifted toward Pyle disease due to the valgus deformity and prominent metaphyseal widening and translucency. Genetic analysis identified 2 pathogenic TNFRSF11A variants: a nonsense mutation c.1093G>T, p.(Glu365*) and a frameshift mutation c.1266_1268delinsCC, p.(Leu422Phefs*104). Thus, genetic and clinical assessment converged on the diagnosis of a mild form of dysosteosclerosis. Both mutations introduced premature stop codons but escaped complete nonsense-mediated decay, potentially permitting residual protein function. Analysis of patient-derived osteoclasts cultured on glass surfaces showed partial differentiation. However, in vitro resorptive function was strongly impaired, which was clinically reflected by reduced serum concentration of the bone resorption marker CTx. Despite this impairment, the retained residual resorptive function likely explains the patient's relatively mild clinical presentation. These findings underscore the complex genetic interactions that affect osteoclast function, leading to a spectrum of phenotypes in osteoclast-related bone disorders.
Collapse
Affiliation(s)
- Dario Gajewski
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Anna Floriane Hennig
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Regina Grün
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Heide Siggelkow
- MVZ Endokrinologikum Göttingen, 37075 Göttingen, Germany
- Department of Trauma, Orthopedics and Reconstructive Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Svenja Vishnolia
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Leonard Bastian
- Department of Orthopedics and Trauma Surgery, Hospital Leverkusen, 51375 Leverkusen, Germany
| | - Hanna Taipaleenmäki
- Institute of Musculoskeletal Medicine, LMU University Hospital, LMU Munich, 82152 Planegg-Martinsried, Germany
- Musculoskeletal University Center Munich, LMU University Hospital, LMU Munich, 82152 Planegg-Martinsried, Germany
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, 89075 Ulm, Germany
| | - Uwe Kornak
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
- MVZ Endokrinologikum Göttingen, 37075 Göttingen, Germany
| | - Eric Hesse
- Institute of Musculoskeletal Medicine, LMU University Hospital, LMU Munich, 82152 Planegg-Martinsried, Germany
- Musculoskeletal University Center Munich, LMU University Hospital, LMU Munich, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
23
|
Yin M, Zheng X, Shi L. Targeting p38 MAPK: A potential bridge between ER stress and age-related bone loss. Cell Signal 2025; 127:111549. [PMID: 39638139 DOI: 10.1016/j.cellsig.2024.111549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The endoplasmic reticulum (ER) is crucial in the development of numerous age-related bone disorders. Notably, ER stress can precipitate bone loss by orchestrating inflammatory responses, apoptosis, and autophagy through the activation of the p38 MAPK pathway. Age-related bone loss diseases pose a significant burden on society and healthcare as the global population ages. This review provides a comprehensive analysis of recent research advancements, delving into the critical role of ER stress-activated p38 MAPK in inflammation, apoptosis, and autophagy, as well as its impact on bone formation and bone resorption. This review elucidates the molecular mechanisms underlying the involvement of ER stress-activated p38 MAPK in osteoporosis, rheumatoid arthritis, periodontitis, and osteoarthritis and discusses the therapeutic potential of targeting p38 MAPK. Furthermore, this review provides a scientific foundation for new therapeutic strategies by highlighting prospective research directions.
Collapse
Affiliation(s)
- Meng Yin
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Zheng
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
24
|
Karadima E, Chavakis T, Alexaki VI. Arginine metabolism in myeloid cells in health and disease. Semin Immunopathol 2025; 47:11. [PMID: 39863828 PMCID: PMC11762783 DOI: 10.1007/s00281-025-01038-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Metabolic flexibility is key for the function of myeloid cells. Arginine metabolism is integral to the regulation of myeloid cell responses. Nitric oxide (NO) production from arginine is vital for the antimicrobial and pro-inflammatory responses. Conversely, the arginase 1 (ARG1)-dependent switch between the branch of NO production and polyamine synthesis downregulates inflammation and promotes recovery of tissue homeostasis. Creatine metabolism is key for energy supply and proline metabolism is required for collagen synthesis. Myeloid ARG1 also regulates extracellular arginine availability and T cell responses in parasitic diseases and cancer. Cancer, surgery, sepsis and persistent inflammation in chronic inflammatory diseases, such as neuroinflammatory diseases or arthritis, are associated with dysregulation of arginine metabolism in myeloid cells. Here, we review current knowledge on arginine metabolism in different myeloid cell types, such as macrophages, neutrophils, microglia, osteoclasts, tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs) and myeloid-derived suppressor cells (MDSCs). A deeper understanding of the function of arginine metabolism in myeloid cells will improve our knowledge on the pathology of several diseases and may set the platform for novel therapeutic applications.
Collapse
Affiliation(s)
- Eleftheria Karadima
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| |
Collapse
|
25
|
Li J, Xiao C, Li C, He J. Tissue-resident immune cells: from defining characteristics to roles in diseases. Signal Transduct Target Ther 2025; 10:12. [PMID: 39820040 PMCID: PMC11755756 DOI: 10.1038/s41392-024-02050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025] Open
Abstract
Tissue-resident immune cells (TRICs) are a highly heterogeneous and plastic subpopulation of immune cells that reside in lymphoid or peripheral tissues without recirculation. These cells are endowed with notably distinct capabilities, setting them apart from their circulating leukocyte counterparts. Many studies demonstrate their complex roles in both health and disease, involving the regulation of homeostasis, protection, and destruction. The advancement of tissue-resolution technologies, such as single-cell sequencing and spatiotemporal omics, provides deeper insights into the cell morphology, characteristic markers, and dynamic transcriptional profiles of TRICs. Currently, the reported TRIC population includes tissue-resident T cells, tissue-resident memory B (BRM) cells, tissue-resident innate lymphocytes, tissue-resident macrophages, tissue-resident neutrophils (TRNs), and tissue-resident mast cells, but unignorably the existence of TRNs is controversial. Previous studies focus on one of them in specific tissues or diseases, however, the origins, developmental trajectories, and intercellular cross-talks of every TRIC type are not fully summarized. In addition, a systemic overview of TRICs in disease progression and the development of parallel therapeutic strategies is lacking. Here, we describe the development and function characteristics of all TRIC types and their major roles in health and diseases. We shed light on how to harness TRICs to offer new therapeutic targets and present burning questions in this field.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
26
|
Sakai E, Tsukuba T. Transcriptomic Characterization Reveals Mitochondrial Involvement in Nrf2/Keap1-Mediated Osteoclastogenesis. Antioxidants (Basel) 2024; 13:1575. [PMID: 39765903 PMCID: PMC11673794 DOI: 10.3390/antiox13121575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Although osteoclasts play crucial roles in the skeletal system, the mechanisms that underlie oxidative stress during osteoclastogenesis remain unclear. The transcription factor Nrf2 and its suppressor, Keap1, function as central mediators of oxidative stress. To further elucidate the function of Nrf2/Keap1-mediated oxidative stress regulation in osteoclastogenesis, DNA microarray analysis was conducted in this study using wild-type (WT), Keap1 knockout (Keap1 KO), and Nrf2 knockout (Nrf2 KO) osteoclasts. Principal component analysis showed that 403 genes, including Nqo1, Il1f9, and Mmp12, were upregulated in Keap1 KO compared with WT osteoclasts, whereas 24 genes, including Snhg6, Ccdc109b, and Wfdc17, were upregulated in Nrf2 KO compared with WT osteoclasts. Moreover, 683 genes, including Car2, Calcr, and Pate4, were upregulated in Nrf2 KO cells compared to Keap1 KO cells. Functional analysis by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis showed upregulated genes in Nrf2 KO osteoclasts were mostly enriched in oxidative phosphorylation. Furthermore, GeneMANIA predicted the protein-protein interaction network of novel molecules such as Rufy4 from genes upregulated in Nrf2 KO osteoclasts. Understanding the complex interactions between these molecules may pave the way for developing promising therapeutic strategies against bone metabolic diseases caused by increased osteoclast differentiation under oxidative stress.
Collapse
Affiliation(s)
- Eiko Sakai
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1, Sakamoto, Nagasaki 852-8588, Japan;
| | | |
Collapse
|
27
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
28
|
Yang H, Tang R, Wu HL, Li JH, Zhang C. Osteoprotective effect of Achyranthes bidentata root extract on osteoporotic rats: a systematic review and meta-analysis. PHARMACEUTICAL BIOLOGY 2024; 62:691-701. [PMID: 39363520 PMCID: PMC11457373 DOI: 10.1080/13880209.2024.2407531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 10/05/2024]
Abstract
CONTEXT Achyranthes bidentata Blume (ABB), a plant of Amaranthaceae family, has been one of the more commonly used phytomedicine remedies for thousands of years, and recent studies have highlighted the efficacy of its extracts in the treatment of osteoporosis. Nonetheless, a thorough analysis of its benefits is currently absent. OBJECTIVE This meta-analysis assessed the effects of ABB root extract (ABBRE) on osteoporotic rats and provides a rationale for future clinical studies. METHODS Searches were conducted in seven different Chinese and English databases, and the search period was from their establishment to January 2024. This study was registered in PROSPERO (CRD42023418917). Selected research regarding the ABBRE treatment of osteoporotic rats, and the corresponding research has distinctly reported outcomes, and the data on the bone mineral density (BMD), bone histomorphometrics, biomechanical parameters, and bone biochemical markers of osteoporotic rats can be extracted. RESULTS Through screening, 11 studies met the eligibility requirements for inclusion, in which 222 animals were studied. The treatment group with ABBRE exhibited increased bone mineral density (standardized mean difference [SMD] = 1.64, 95% CI = 0.52 to 2.77). Based on subgroup analysis, the greatest increase in bone mineral density was observed when the dose of ABBRE was ≤ 400 mg/kg/day and the duration of treatment was ≤ 12 weeks. CONCLUSIONS ABBRE is a phytomedicine that can effectively promote the enhancement of bone mineral density and ease osteoporosis. It can be developed into a new alternative therapy by conducting experiments and clinical studies on larger samples.
Collapse
Affiliation(s)
- Hao Yang
- The Clinical Medical College, Chengdu University of Chinese Medicine, Chengdu City, Sichuan Province, China
| | - Rui Tang
- The Clinical Medical College, Chengdu University of Chinese Medicine, Chengdu City, Sichuan Province, China
| | - Hong-Li Wu
- The Clinical Medical College, Chengdu University of Chinese Medicine, Chengdu City, Sichuan Province, China
| | - Jia-Hao Li
- The Clinical Medical College, Chengdu University of Chinese Medicine, Chengdu City, Sichuan Province, China
| | - Chi Zhang
- The Health Preservation and Rehabilitation College, Chengdu University of Chinese Traditional Medicine, Chengdu City, Sichuan Province, China
| |
Collapse
|
29
|
Zhang X, Feng C, Yuan T, Wang Y, Wang H, Lu Q, Lv Y, Li Z, Fu C, Sun S. Inhibition of protein disulfide isomerase mitigates steroid-induced osteonecrosis of the femoral head by suppressing osteoclast activity through the reduction of cellular oxidative stress. Chem Biol Interact 2024; 404:111263. [PMID: 39393751 DOI: 10.1016/j.cbi.2024.111263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/21/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Osteonecrosis of the femoral head (ONFH) is a devastating and irreversible hip disease usually associated with increased oxidative stress due to the clinical application of high-dose or long-term glucocorticoids (GCs). Previous publications have demonstrated protein disulfide isomerase (PDI) plays a critical role in regulating cellular production of reactive oxygen species (ROS). We therefore ask whether interfering PDI could affect GCs-stimulated osteoclastogenesis. To test the hypothesis, we conducted bioinformatics and network analysis based on potential gene targets of steroid-induced osteonecrosis of the femoral head (SIONFH) in light of multiple databases and concomitantly verified the associated biological effect via the in vitro model of dexamethasone (DEX)-stimulated osteoclastogenesis. The results revealed 70 potential gene targets for SIONFH intervention, including the P4HB gene that encodes PDI. Further analysis based on network topology-based analysis techniques (NTA), protein-protein interaction (PPI) networks, and mouse cell atlas database identified the importance of PDI in regulating the cellular redox state of osteoclast during ONFH. Western blotting (WB) validations also indicated that PDI may be a positive regulator in the process of DEX-stimulated osteoclastogenesis. Hence, various PDI inhibitors were subjected to molecular docking with PDI and their performances were analyzed, including 3-Methyltoxoflavin (3 M) which inhibits PDI expression, and ribostamycin sulfate (RS) which represses PDI chaperone activity. The binding energies of DEX, 3 M, and RS to PDI were -5.3547, -4.2324, and -5.9917 kcal/mol, respectively. The Protein-Ligand Interaction Profiler (PLIP) analysis demonstrated that both hydrogen bonds and hydrophobic interactions were the key contributions to the DEX-PDI and 3M-PDI complexes, while only hydrogen bonds were identified as the predominant driving forces in the RS-PDI complex. Subsequent experiments showed that both 3 M and RS reduced osteoclast differentiation and bone resorption activity by stifling the expression of osteoclastic markers. This reduction was primarily due to the PDI inhibitors boosting the antioxidant system, thereby reducing the production of intracellular ROS. In conclusion, our results supported PDI's involvement in SIONFH progression by regulating ROS in osteoclasts and highlighted PDI inhibitors may serve as potential options for SIONFH treatment.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Changgong Feng
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Tao Yuan
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yi Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Qizhen Lu
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - YongShuang Lv
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Ziqing Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Chuanyun Fu
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
30
|
Peng Y, Wang Y, Bai R, Shi K, Zhou H, Chen C. Nanomaterials: Recent Advances in Knee Osteoarthritis Treatment. Adv Healthc Mater 2024; 13:e2400615. [PMID: 39308252 DOI: 10.1002/adhm.202400615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/16/2024] [Indexed: 12/28/2024]
Abstract
Osteoarthritis (OA) of the knee is the most prevalent degenerative joint condition that places a substantial financial and medical burden on society. However, due to drawbacks such as inefficiency, adverse effects, and brief duration of action, the clinical efficacy of the current major therapies for knee OA is largely restricted. Therefore, novel medication development is highly required to address these issues. Numerous studies in recent years have established that nanomaterials can be a potential and highly effective way to overcome these challenges. In this review, the anatomical distinctions between healthy and OA knee joints, as well as novel advances in the field of nanomaterials for the treatment of knee OA are summarized. The limits of the present therapeutic strategies for treating knee OA are also highlighted, as well as the potential prospects of nanomaterials in the future.
Collapse
Affiliation(s)
- Yufeng Peng
- Henan Institutes of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, China
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
| | - Ying Wang
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Ru Bai
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Kejian Shi
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Huige Zhou
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Chunying Chen
- Henan Institutes of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, China
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing, 100021, China
| |
Collapse
|
31
|
Hong S, Kwon J, Song S, Park I, Jung DS, Saruul E, Nho CW, Kwon HC, Yoo G. Suppressive Effects of Geoje Raspberry ( Rubus tozawae Nakai ex J.Y. Yang) on Post-Menopausal Osteoporosis via Its Osteogenic Activity on Osteoblast Differentiation. Nutrients 2024; 16:3856. [PMID: 39599642 PMCID: PMC11597101 DOI: 10.3390/nu16223856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/01/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Osteoporosis is a metabolic bone disease with a high mortality rate due to non-traumatic fractures. The risk of osteoporosis is increasing globally due to an increasing aging population. Current therapies are limited to delaying disease progression. Recently, the need to discover foods with osteogenic activity for the prevention and treatment of osteoporosis has been emphasized. We focused on bone formation via osteoblast differentiation, considering bone formation and resorption during bone homeostasis. Rubus tozawae Nakai ex J. Y. Yang (RL, Geoje raspberry) is a deciduous subshrub that has been traditionally eaten for its fruit. METHODS AND RESULTS We identified the third subfraction of n-hexane fraction (RL-Hex-NF3) of RL, an endemic Korean plant with osteogenic activity, which increased bone density in ovariectomized mice, a representative animal model of osteoporosis, via the depletion of female hormones, which resulted from the increase in the osteoblast population. RL-Hex-NF3 induced osteoblast differentiation and the expression of osteogenic markers in MC3T3-E1 pre-osteoblasts. Seven compounds were identified from RL-Hex-NF3 using NMR spectroscopy. Of these, three compounds, namely, 3β-hydroxy-18α,19α-urs-20-en-28-oic acid, betulinic acid, and (1S,6R,7S)-muurola-4,10(14)-diene-15-ol, showed strong osteogenic activity. CONCLUSIONS RL-Hex-NF3 and its compounds suppress bone loss via their osteogenic properties, suggesting that they could be a potent candidate to treat osteoporosis.
Collapse
Affiliation(s)
- Soyeon Hong
- Smart Farm Research Center, Korean Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (S.H.); (D.S.J.); (E.S.); (C.W.N.)
| | - Jaeyoung Kwon
- Natural Product Informatics Research Center, Korean Institute of Science and Technology, Gangneung 25451, Republic of Korea; (J.K.); (S.S.); (I.P.)
- Department of Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Sungmin Song
- Natural Product Informatics Research Center, Korean Institute of Science and Technology, Gangneung 25451, Republic of Korea; (J.K.); (S.S.); (I.P.)
| | - InWha Park
- Natural Product Informatics Research Center, Korean Institute of Science and Technology, Gangneung 25451, Republic of Korea; (J.K.); (S.S.); (I.P.)
| | - Da Seul Jung
- Smart Farm Research Center, Korean Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (S.H.); (D.S.J.); (E.S.); (C.W.N.)
| | - Erdenebileg Saruul
- Smart Farm Research Center, Korean Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (S.H.); (D.S.J.); (E.S.); (C.W.N.)
| | - Chu Won Nho
- Smart Farm Research Center, Korean Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (S.H.); (D.S.J.); (E.S.); (C.W.N.)
- Department of Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hak Cheol Kwon
- Natural Product Informatics Research Center, Korean Institute of Science and Technology, Gangneung 25451, Republic of Korea; (J.K.); (S.S.); (I.P.)
| | - Gyhye Yoo
- Smart Farm Research Center, Korean Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (S.H.); (D.S.J.); (E.S.); (C.W.N.)
- Department of Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
32
|
Gal M, Tuan HM, Park JH, Park KH, Kim O, Min BS, Lee JH. Irilin D suppresses RANKL-induced osteoclastogenesis and prevents inflammation-induced bone loss by disrupting the NF-κB and MAPK signaling pathways. Eur J Pharmacol 2024; 982:176956. [PMID: 39209096 DOI: 10.1016/j.ejphar.2024.176956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Excessive activity of osteoclasts(OCs) lead to bone resorption in chronic inflammatory conditions. The use of natural compounds to target OCs offers significant promise in the treatment or prevention of OC-associated diseases. Irilin D (IRD), a natural isoflavone derived from Belamcanda chinensis (L.) DC., has potential effects on OC differentiation both in vitro and in vivo that have yet to be thoroughly explored. In our study, we found that IRD inhibited receptor activator of nuclear factor-κB ligand (RANKL)-induced OC differentiation, actin ring formation, and bone resorption in vitro without compromising cell viability. However, IRD did not exhibit anti-inflammatory effects in lipopolysaccharide (LPS)-stimulated macrophages. Furthermore, IRD reduced LPS-induced inflammatory bone loss by blocking osteoclastogenesis in a mouse model. Mechanistically, IRD disrupted RANKL-induced activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB), leading to the inhibition of c-Fos and nuclear factor of activated T cells cytoplasmic 1 (NFATc1) activation. We also demonstrated that IRD inhibited RANKL-induced osteoclastic NFATc1 target genes, including DC-STAMP, ACP5, and CtsK. Our results indicate that IRD mitigates LPS-induced inflammatory bone resorption in mice by inhibiting RANKL-activated MAPKs and NF-κB signaling pathways, suggesting its potential as a natural isoflavone for preventing or treating OC-associated diseases.
Collapse
Affiliation(s)
- Minju Gal
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea
| | - Ha Manh Tuan
- College of Pharmacy, Drug Research and Development Center, Daegu Catholic University, Gyeongbuk, 38430, Republic of Korea
| | - Ju-Hee Park
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea
| | - Kang-Hyeon Park
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea
| | - Okhwa Kim
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea
| | - Byung-Sun Min
- College of Pharmacy, Drug Research and Development Center, Daegu Catholic University, Gyeongbuk, 38430, Republic of Korea.
| | - Jeong-Hyung Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea; Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea.
| |
Collapse
|
33
|
Indarwulan N, Savitri M, Ashariati A, Bintoro SUY, Diansyah MN, Amrita PNA, Romadhon PZ. Bone Mineral Density, C-Terminal Telopeptide of Type I Collagen, and Osteocalcin as Monitoring Parameters of Bone Remodeling in CML Patients Undergoing Imatinib Therapy: A Basic Science and Clinical Review. Diseases 2024; 12:275. [PMID: 39589949 PMCID: PMC11592756 DOI: 10.3390/diseases12110275] [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: 09/17/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Chronic myeloid leukemia (CML) is one of the most commonly found types of myeloproliferative neoplasms, characterized by increased proliferation of granulocytic cells without losing their differentiation ability. Imatinib, a tyrosine kinase inhibitor (TKI), can be effectively used as therapy for CML. However, Imatinib can affect bone turnover thus having clinical implications on the bones of CML patients undergoing long-term Imatinib therapy. However, parameters that can accurately describe the bone condition in CML patients receiving Imatinib still need further study. A combination of imaging techniques such as bone mineral density (BMD) and bone turnover activity markers such as C-terminal telopeptide of type I collagen (CTX-1) and osteocalcin has the potential to be used as monitoring parameters for bone density abnormalities in CML patients receiving Imatinib. OBJECTIVES This article explains the rationale for using BMD, CTX-1, and osteocalcin as monitoring parameters of bone remodeling in CML patients receiving Imatinib. RESULTS First, the physiological process of bone turnover will be explained. Then, we describe the role of tyrosine kinase in bone metabolism. Next, the impact of Imatinib on BMD, CTX-1, and osteocalcin will be explained. CONCLUSION The assessment of bone health of CML patients on Imatinib should include both BMD tests and bone turnover marker assays such as CTX-1 and osteocalcin.
Collapse
Affiliation(s)
- Nurita Indarwulan
- Subspeciality Program in Hematology and Medical Oncology Division, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia;
- Subspeciality Program in Hematology and Medical Oncology Division, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Merlyna Savitri
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Ami Ashariati
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Siprianus Ugroseno Yudho Bintoro
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Muhammad Noor Diansyah
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Putu Niken Ayu Amrita
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Pradana Zaky Romadhon
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| |
Collapse
|
34
|
Zheng Y, Li J, Li Y, Wang J, Suo C, Jiang Y, Jin L, Xu K, Chen X. Plasma proteomic profiles reveal proteins and three characteristic patterns associated with osteoporosis: A prospective cohort study. J Adv Res 2024:S2090-1232(24)00474-0. [PMID: 39490735 DOI: 10.1016/j.jare.2024.10.019] [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: 06/15/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
INTRODUCTION Exploration of plasma proteins associated with osteoporosis can offer insights into its pathological development, identify novel biomarkers for screening high-risk populations, and facilitate the discovery of effective therapeutic targets. OBJECTIVES The present study aimed to identify potential proteins associated with osteoporosis and to explore the underlying mechanisms from a proteomic perspective. METHODS The study included 42,325 participants without osteoporosis in the UK Biobank (UKB), of whom 1,477 developed osteoporosis during the follow-up. We used Cox regression and Mendelian randomization analysis to examine the association between plasma proteins and osteoporosis. Machine learning was utilized to explore proteins with strong predictive power for osteoporosis risk. RESULTS Of 2,919 plasma proteins, we identified 134 significantly associated with osteoporosis, with sclerostin (SOST), adiponectin (ADIPOQ), and creatine kinase B-type (CKB) exhibiting strong associations. Twelve of these proteins showed significant associations with bone mineral density (BMD) T-score at the femoral neck, lumbar spine, and total body. Mendelian randomization further supported causal relationships between 17 plasma proteins and osteoporosis. Moreover, follitropin subunit beta (FSHB), SOST, and ADIPOQ demonstrated high importance in predictive modeling. Utilizing a predictive model built with 10 proteins, we achieved relatively accurate prediction of osteoporosis onset up to 5 years in advance (AUC = 0.803). Finally, we identified three osteoporosis-related protein modules associated with immunity, lipid metabolism, and follicle-stimulating hormone (FSH) regulation from a network perspective, elucidating their mediating roles between various risk factors (smoking, sleep, physical activity, polygenic risk score (PRS), and menopause) and osteoporosis. CONCLUSION We identified several proteins associated with osteoporosis and highlighted the role of plasma proteins in influencing its progression through three primary pathways: immunity, lipid metabolism, and FSH regulation. This provides further insights into the distinct molecular patterns and pathogenesis of bone loss and may contribute to strengthening early diagnosis and long-term monitoring of the condition.
Collapse
Affiliation(s)
- Yi Zheng
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Jincheng Li
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yucan Li
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Jiacheng Wang
- Department of Epidemiology, School of Public Health, and the Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Chen Suo
- Department of Biostatistics, School of Public Health, and the Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Kelin Xu
- Department of Biostatistics, School of Public Health, and the Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China.
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, China.
| |
Collapse
|
35
|
Liang Y, Qin T, Pang C, Yang X, Wu Z, Liao X, Zhang J, Zeng S, Zhou C, Liu C. Chongrenside D from Smilax china L protects against inflammation-induced joint destruction via inhibiting osteoclastogenesis. Heliyon 2024; 10:e38818. [PMID: 39430543 PMCID: PMC11490767 DOI: 10.1016/j.heliyon.2024.e38818] [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: 01/30/2024] [Revised: 09/06/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024] Open
Abstract
Background Bone-destructive diseases including rheumatoid arthritis (RA), osteoporosis, and bone metastases, are increasingly prevalent and worrisome due to the over-activated of osteoclasts. Chongrenside D (CGD) is a furostanol saponin extracted from Smilax china L, which has been demonstrated to have anti-inflammatory properties in our previous research. However, its effect on rheumatoid arthritis, especially on osteoclast differentiation and bone destruction has not yet been investigated. Methods We evaluated the toxicity of CGD on the cell we used, RANKL-induced osteoclast formation, bone resorption activity, and osteoclast-specific genes or protein expression using bone marrow-derived monocytes (BMMs) -derived osteoclasts. Furthermore, the protective function of CGD on the paws of osteolytic mice was carried out using micro-CT, H&E, TRAP staining, as well as real-time PCR, and western blotting. Inflammatory cytokine levels were conducted through ELISA assay. The relative signaling pathways were investigated using western blotting, immunofluorescence microscopy and real-time PCR. Results CGD notably inhibited RANKL-induced osteoclast formation, and suppressed the expression of osteoclast markers and actin ring formation, thus attenuating its bone resorption activity. For in vivo work, CGD protected against joint bone destruction induced by LPS, increased trabecular number and thickness, and reduced trabecular separation. CGD also inhibited the levels of inflammatory cytokines IL-6and TNF-α, improved the integrity of joint bones and decreased TRAP-positive staining area. The mechanistic study indicated that CGD down-regulated MMP9 and FAK-Src signaling, which were crucial for the resorption function of osteoclasts. CGD also inhibited MAPK pathway-mediated cell differentiation and survival, finally resulting in weak osteoclastogenesis. Conclusion CGD exerts a significant anti-osteolytic activity both in vitro and in vivo by inhibiting RANKL-induced osteoclastogenesis and function. Consequently, our study indicated that CGD may have a potential therapeutic role in the precaution of osteolytic bone disease.
Collapse
Affiliation(s)
- Yanxiang Liang
- Department of Pharmacy, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, 518101, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
| | - Tian Qin
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
| | - Caixia Pang
- Department of Pharmacy, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, 518101, China
| | - Xinru Yang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
| | - Zongbin Wu
- Department of Pharmacy, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, 518101, China
| | - Xiaolian Liao
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
| | - Jie Zhang
- Department of Pharmacy, Guangdong Provincial Hospital of Chinese Medicine. the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 111 Dade Road, Guangzhou, 510120, China
| | - Siyu Zeng
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Chun Zhou
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
| | - Cuiling Liu
- Department of Pharmacy, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, 518101, China
| |
Collapse
|
36
|
Bertels JC, He G, Long F. Metabolic reprogramming in skeletal cell differentiation. Bone Res 2024; 12:57. [PMID: 39394187 PMCID: PMC11470040 DOI: 10.1038/s41413-024-00374-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/13/2024] Open
Abstract
The human skeleton is a multifunctional organ made up of multiple cell types working in concert to maintain bone and mineral homeostasis and to perform critical mechanical and endocrine functions. From the beginning steps of chondrogenesis that prefigures most of the skeleton, to the rapid bone accrual during skeletal growth, followed by bone remodeling of the mature skeleton, cell differentiation is integral to skeletal health. While growth factors and nuclear proteins that influence skeletal cell differentiation have been extensively studied, the role of cellular metabolism is just beginning to be uncovered. Besides energy production, metabolic pathways have been shown to exert epigenetic regulation via key metabolites to influence cell fate in both cancerous and normal tissues. In this review, we will assess the role of growth factors and transcription factors in reprogramming cellular metabolism to meet the energetic and biosynthetic needs of chondrocytes, osteoblasts, or osteoclasts. We will also summarize the emerging evidence linking metabolic changes to epigenetic modifications during skeletal cell differentiation.
Collapse
Affiliation(s)
- Joshua C Bertels
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Guangxu He
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Orthopedics, The Second Xiangya Hospital, Changsha, Hunan, China
| | - Fanxin Long
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Wang L, Wang S, Dai X, Yue G, Yin J, Xu T, Shi H, Liu T, Jia Z, Brömme D, Zhang S, Zhang D. Salvia miltiorrhiza in osteoporosis: a review of its phytochemistry, traditional clinical uses and preclinical studies (2014-2024). Front Pharmacol 2024; 15:1483431. [PMID: 39421672 PMCID: PMC11484006 DOI: 10.3389/fphar.2024.1483431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Osteoporosis becomes a global public health concern due to its rising prevalence and substantial impact on life quality. Salvia miltiorrhiza Bunge (Salviae Miltiorrhizae Radix et Rhizoma, SM) has been firstly recorded in Shen Nong's Herbal Classic, and is frequently prescribed in conjunction with other herbs for the management of osteoporosis. This systematic review aims to comprehensively analyze the recent advances of SM on osteoporosis in traditional Chinese clinical uses and preclinical investigations. Literature encompassing pertinent studies were systematically retrieved across multiple databases, including the PubMed, Web of Science, Chinese National Knowledge Infrastructure, Chinese VIP Database, and Chinese Biomedical Literature Database. Original investigations spanning from February 2014 to March 2024, including traditional Chinese medicine (TCM) clinical trials and preclinical studies, were employed to analyze the effects and actions of SM on osteoporosis. Thirty-eight TCM clinical trials were identified to employ SM in combination with other herbs for the management of primary and secondary osteoporosis. The overall efficacy was between 77% and 96.67%. Forty preclinical studies were identified to investigate the effects and actions of SM and/or its ingredients on osteoporosis. The anti-osteoporosis actions of this herb may be attributed to inhibit osteoclastogenesis/bone resorption and promote osteoblastogenesis/osteogenesis. The ethanol extracts and its ingredients (tanshinones) inhibit osteoclastogenesis/bone resorption by inhibiting the MAPK/NF-κB/NFATc1 signaling pathway and cathepsin K-induced collagen degradation. Both ethanol extracts (tanshinones) and water extracts (Sal B and tanshinol) contribute to osteoblastogenesis by promoting osteogenesis and angiogenesis via activation of the Wnt/β-catenin/VEGF and ERK/TAZ pathways, and eliminating ROS production targeting Nrf2/ARE/HO-1 pathway. In conclusions, SM may offer a novel strategy for osteoporosis management. Well-designed clinical trials are still needed to evaluate the actions of this herb and its ingredients on bone remodeling.
Collapse
Affiliation(s)
- Lili Wang
- Department of TCM Pharmacology, Chinese Material Medica School, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Wang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xuan Dai
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Gaiyue Yue
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jiyuan Yin
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tianshu Xu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hanfen Shi
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tianyuan Liu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhanhong Jia
- Department of TCM Pharmacology, Chinese Material Medica School, Beijing University of Chinese Medicine, Beijing, China
| | - Dieter Brömme
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, Canada
| | - Shuofeng Zhang
- Department of TCM Pharmacology, Chinese Material Medica School, Beijing University of Chinese Medicine, Beijing, China
| | - Dongwei Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
38
|
Xu C, Wei Z, Lv L, Dong X, Xia W, Xing J, Liu H, Zhao X, Liu Y, Wang W, Jiang H, Gong Y, Liu C, Xu K, Wang S, Akimoto Y, Hu Z. Impdh2 deficiency suppresses osteoclastogenesis through mitochondrial oxidative phosphorylation and alleviates ovariectomy-induced osteoporosis. Biochem Biophys Res Commun 2024; 727:150317. [PMID: 38959733 DOI: 10.1016/j.bbrc.2024.150317] [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: 06/17/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Abnormalities in osteoclastic generation or activity disrupt bone homeostasis and are highly involved in many pathologic bone-related diseases, including rheumatoid arthritis, osteopetrosis, and osteoporosis. Control of osteoclast-mediated bone resorption is crucial for treating these bone diseases. However, the mechanisms of control of osteoclastogenesis are incompletely understood. In this study, we identified that inosine 5'-monophosphate dehydrogenase type II (Impdh2) positively regulates bone resorption. By histomorphometric analysis, Impdh2 deletion in mouse myeloid lineage cells (Impdh2LysM-/- mice) showed a high bone mass due to the reduced osteoclast number. qPCR and western blotting results demonstrated that the expression of osteoclast marker genes, including Nfatc1, Ctsk, Calcr, Acp5, Dcstamp, and Atp6v0d2, was significantly decreased in the Impdh2LysM-/- mice. Furthermore, the Impdh inhibitor MPA treatment inhibited osteoclast differentiation and induced Impdh2-cytoophidia formation. The ability of osteoclast differentiation was recovered after MPA deprivation. Interestingly, genome-wide analysis revealed that the osteoclastic mitochondrial biogenesis and functions, such as oxidative phosphorylation, were impaired in the Impdh2LysM-/- mice. Moreover, the deletion of Impdh2 alleviated ovariectomy-induced bone loss. In conclusion, our findings revealed a previously unrecognized function of Impdh2, suggesting that Impdh2-mediated mechanisms represent therapeutic targets for osteolytic diseases.
Collapse
Affiliation(s)
- Cheng Xu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China.
| | - Zhixin Wei
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei, 430056, China
| | - Longfei Lv
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei, 430056, China
| | - Xiaoyu Dong
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei, 430056, China
| | - Wenwen Xia
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Junqiao Xing
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Hongni Liu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Xue Zhao
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Yuan Liu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Weihua Wang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Haochen Jiang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Yeli Gong
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China
| | - Cong Liu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Kai Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Siyuan Wang
- Department of Medicinal Chemistry, College of Pharmacy, Shenzhen Technology University, Shenzhen, Guangdong, 518118, China
| | | | - Zhangfeng Hu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China; Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei, 430056, China; Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Jianghan University, Wuhan, Hubei, 430056, China.
| |
Collapse
|
39
|
Campbell MJ, Bustamante-Gomez C, Fu Q, Beenken KE, Reyes-Pardo H, Smeltzer MS, O'Brien CA. RANKL-mediated osteoclast formation is required for bone loss in a murine model of Staphylococcus aureus osteomyelitis. Bone 2024; 187:117181. [PMID: 38960295 PMCID: PMC11325436 DOI: 10.1016/j.bone.2024.117181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Staphylococcus aureus osteomyelitis leads to extensive bone destruction. Osteoclasts are bone resorbing cells that are often increased in bone infected with S. aureus. The cytokine RANKL is essential for osteoclast formation under physiological conditions but in vitro evidence suggests that inflammatory cytokines may by-pass the requirement for RANKL. The goal of this study was to determine whether RANKL-dependent osteoclast formation is essential for the bone loss that occurs in a murine model of S. aureus osteomyelitis. To this end, humanized-RANKL mice were infected by direct inoculation of S. aureus into a unicortical defect in the femur. Mice were treated with vehicle or denosumab, a human monoclonal antibody that inhibits RANKL, both before and during a 14-day infection period. The severe cortical bone destruction caused by infection was completely prevented by denosumab administration even though the bacterial burden in the femur was not affected. Osteoclasts were abundant near the inoculation site in vehicle-treated mice but absent in denosumab-treated mice. In situ hybridization demonstrated that S. aureus infection potently stimulated RANKL expression in bone marrow stromal cells. The extensive reactive bone formation that occurs in this osteomyelitis model was also reduced by denosumab administration. Lastly, there was a notable lack of osteoblasts near the infection site suggesting that the normal coupling of bone formation to bone resorption was disrupted by S. aureus infection. These results demonstrate that RANKL-mediated osteoclast formation is required for the bone loss that occurs in S. aureus infection and suggest that disruption of the coupling of bone formation to bone resorption may also contribute to bone loss in this condition.
Collapse
Affiliation(s)
- Mara J Campbell
- Department of Microbiology and Immunology, The University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Cecile Bustamante-Gomez
- Division of Endocrinology, The University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Qiang Fu
- Division of Endocrinology, The University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Karen E Beenken
- Department of Microbiology and Immunology, The University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Humberto Reyes-Pardo
- Division of Endocrinology, The University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Mark S Smeltzer
- Department of Microbiology and Immunology, The University of Arkansas for Medical Sciences, Little Rock, AR, United States of America; Department of Orthopaedic Surgery, The University of Arkansas for Medical Sciences, Little Rock, AR, United States of America.
| | - Charles A O'Brien
- Division of Endocrinology, The University of Arkansas for Medical Sciences, Little Rock, AR, United States of America; Department of Orthopaedic Surgery, The University of Arkansas for Medical Sciences, Little Rock, AR, United States of America; Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America.
| |
Collapse
|
40
|
Chu W, Peng W, Lu Y, Liu Y, Li Q, Wang H, Wang L, Zhang B, Liu Z, Han L, Ma H, Yang H, Han C, Lu X. PRMT6 Epigenetically Drives Metabolic Switch from Fatty Acid Oxidation toward Glycolysis and Promotes Osteoclast Differentiation During Osteoporosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403177. [PMID: 39120025 PMCID: PMC11516099 DOI: 10.1002/advs.202403177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/23/2024] [Indexed: 08/10/2024]
Abstract
Epigenetic regulation of metabolism profoundly influences cell fate commitment. During osteoclast differentiation, the activation of RANK signaling is accompanied by metabolic reprogramming, but the epigenetic mechanisms by which RANK signaling induces this reprogramming remain elusive. By transcriptional sequence and ATAC analysis, this study identifies that activation of RANK signaling upregulates PRMT6 by epigenetic modification, triggering a metabolic switching from fatty acids oxidation toward glycolysis. Conversely, Prmt6 deficiency reverses this shift, markedly reducing HIF-1α-mediated glycolysis and enhancing fatty acid oxidation. Consequently, PRMT6 deficiency or inhibitor impedes osteoclast differentiation and alleviates bone loss in ovariectomized (OVX) mice. At the molecular level, Prmt6 deficiency reduces asymmetric dimethylation of H3R2 at the promoters of genes including Ppard, Acox3, and Cpt1a, enhancing genomic accessibility for fatty acid oxidation. PRMT6 thus emerges as a metabolic checkpoint, mediating metabolic switch from fatty acid oxidation to glycolysis, thereby supporting osteoclastogenesis. Unveiling PRMT6's critical role in epigenetically orchestrating metabolic shifts in osteoclastogenesis offers a promising target for anti-resorptive therapy.
Collapse
Affiliation(s)
- Wenxiang Chu
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Weilin Peng
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Yingying Lu
- Obstetrics and Gynecology HospitalFudan UniversityShanghai200011China
| | - Yishan Liu
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Qisheng Li
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Haibin Wang
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Liang Wang
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Bangke Zhang
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Zhixiao Liu
- Histology and Embryology Department and Shanghai Key Laboratory of Cell EngineeringNaval Medical UniversityShanghai200433China
| | - Lin Han
- Department of OrthopaedicsThird Affiliated Hospital of Naval Medical UniversityShanghai201805China
| | - Hongdao Ma
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Haisong Yang
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Chaofeng Han
- Histology and Embryology Department and Shanghai Key Laboratory of Cell EngineeringNaval Medical UniversityShanghai200433China
- National Key Laboratory of Immunity and Inflammation, Institute of ImmunologyNaval Medical UniversityShanghai200433China
| | - Xuhua Lu
- Department of Orthopaedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| |
Collapse
|
41
|
Marie Encarnacion A, Pootheri N, Yao H, Chen Z, Lee S, Kim E, Lee TH. Novel inhibitor N-cyclopropyl-4-((4-((4-(trifluoromethyl)phenyl)sulfonyl)piperazin-1-yl)methyl)benzamide attenuates RANKL-mediated osteoclast differentiation in vitro. Bioorg Med Chem Lett 2024; 110:129884. [PMID: 38996939 DOI: 10.1016/j.bmcl.2024.129884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/24/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Both cyclopropyl amide and piperazine sulfonamide functional groups are known for their various biological properties used for drug development. Herein, we synthesized nine new derivatives with different substituent groups incorporating these moieties and screened them for their anti-osteoclast differentiation activity. After analyzing the structure-activity relationship (SAR), the inhibitory effect against osteoclastogenesis was determined to be dependent on the lipophilicity of the compound. Derivative 5b emerged as the most effective dose-dependent inhibitor after TRAP staining with an IC50 of 0.64 µM against RANKL-induced osteoclast cells. 5b was also able to suppress F-acting ring formation and bone resorption activity of osteoclasts in vitro. Finally, well-acknowledged gene and protein osteoclast-specific marker expression levels were decreased after 5b administration on primary murine osteoclast cells.
Collapse
Affiliation(s)
| | - Nithin Pootheri
- Department of Chemistry, Chonnam National University, Gwangju 61186, South Korea
| | - Hongyuan Yao
- Interdisciplinary Department of Biomedical Engineering, Chonnam National University, Gwangju 61186, South Korea
| | - Zhihao Chen
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, South Korea
| | - Sunwoo Lee
- Department of Chemistry, Chonnam National University, Gwangju 61186, South Korea
| | - Eunae Kim
- College of Pharmacy, Chosun University, Gwangju 61452, South Korea; Host-directed Antiviral Research Center, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Tae-Hoon Lee
- Interdisciplinary Department of Biomedical Engineering, Chonnam National University, Gwangju 61186, South Korea; Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
42
|
Chu F, Wang Z, Zhang D, Xu W, Huang B, Long C, Yang S, Qu X, Gao C, Yuan F. Research on the osteogenic properties of 3D-printed porous titanium alloy scaffolds loaded with Gelma/PAAM-ZOL composite hydrogels. Int J Biol Macromol 2024; 276:134050. [PMID: 39038567 DOI: 10.1016/j.ijbiomac.2024.134050] [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: 04/10/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Although titanium alloy is the most widely used endoplant material in orthopedics, the material is bioinert and good bone integration is difficult to achieve. Zoledronic acid (ZOL) has been shown to locally inhibit osteoclast formation and prevent osteoporosis, but excessive concentrations of ZOL exert an inhibitory effect on osteoblasts; therefore, stable and controlled local release of ZOL may reshape bone balance and promote bone regeneration. To promote the adhesion of osteoblasts to many polar groups, researchers have applied gelatine methacryloyl (Gelma) combined with polyacrylamide hydrogel (PAAM), which significantly increased the hydrogen bonding force between the samples and improved the stability of the coating and drug release. A series of experiments demonstrated that the Gelma/PAAM-ZOL bioactive coating on the surface of the titanium alloy was successfully prepared. The coating can induce osteoclast apoptosis, promote osteoblast proliferation and differentiation, achieve dual regulation of bone regeneration, successfully disrupt the balance of bone remodelling and promote bone tissue regeneration. Additionally, the coating improves the metal biological inertness on the surface of titanium alloys and improves the bone integration of the scaffold, offering a new strategy for bone tissue engineering to promote bone technology.
Collapse
Affiliation(s)
- Fuchao Chu
- Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China; Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China
| | - Zhenxin Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China
| | - Dazhen Zhang
- Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China
| | - Wenkang Xu
- Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China; Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China
| | - Boyan Huang
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China
| | - Chen Long
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China
| | - Shuo Yang
- Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China; Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China
| | - Xinzhe Qu
- Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China
| | - Cunjiu Gao
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China
| | - Feng Yuan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, Jiangsu, , China.
| |
Collapse
|
43
|
Deng D, Liu X, Huang W, Yuan S, Liu G, Ai S, Fu Y, Xu H, Zhang X, Li S, Xu S, Bai X, Zhang Y. Osteoclasts control endochondral ossification via regulating acetyl-CoA availability. Bone Res 2024; 12:49. [PMID: 39198395 PMCID: PMC11358419 DOI: 10.1038/s41413-024-00360-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/27/2024] [Accepted: 07/21/2024] [Indexed: 09/01/2024] Open
Abstract
Osteoclast is critical in skeletal development and fracture healing, yet the impact and underlying mechanisms of their metabolic state on these processes remain unclear. Here, by using osteoclast-specific small GTPase Rheb1-knockout mice, we reveal that mitochondrial respiration, rather than glycolysis, is essential for cathepsin K (CTSK) production in osteoclasts and is regulated by Rheb1 in a mechanistic target of rapamycin complex 1 (mTORC1)-independent manner. Mechanistically, we find that Rheb1 coordinates with mitochondrial acetyl-CoA generation to fuel CTSK, and acetyl-CoA availability in osteoclasts is the central to elevating CTSK. Importantly, our findings demonstrate that the regulation of CTSK by acetyl-CoA availability is critical and may confer a risk for abnormal endochondral ossification, which may be the main cause of poor fracture healing on alcohol consumption, targeting Rheb1 could successfully against the process. These findings uncover a pivotal role of mitochondria in osteoclasts and provide a potent therapeutic opportunity in bone disorders.
Collapse
Affiliation(s)
- Daizhao Deng
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xianming Liu
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Wenlan Huang
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Sirui Yuan
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Genming Liu
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Shanshan Ai
- Department of Physiology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yijie Fu
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Haokun Xu
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xinyi Zhang
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Shihai Li
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Song Xu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Yue Zhang
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
44
|
Qin L, Lu J, He Q, Wang H, Yao L, Duffy M, Guo H, Braun C, Lin Y, Zhou Y, Liang Q, Bandyopadhyay S, Tan K, Choi Y, Liu S. Bone marrow adipogenic lineage precursors are the major regulator of bone resorption in adult mice. RESEARCH SQUARE 2024:rs.3.rs-4809633. [PMID: 39257979 PMCID: PMC11384808 DOI: 10.21203/rs.3.rs-4809633/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Bone resorption by osteoclasts is a critical step in bone remodeling, a process important for maintaining bone homeostasis and repairing injured bone. We previously identified a bone marrow mesenchymal subpopulation, marrow adipogenic lineage precursors (MALPs), and showed that its production of RANKL stimulates bone resorption in young mice using Adipoq-Cre. To exclude developmental defects and to investigate the role of MALPs-derived RANKL in adult bone, we generated inducible reporter mice (Adipoq-CreER Tomato) and RANKL deficient mice (Adipoq-CreER RANKLflox/flox, iCKO). Single cell-RNA sequencing data analysis, lineage tracing, and in situ hybridization revealed that Adipoq+ cells contain not only MALPs but also late mesenchymal progenitors capable of osteogenic differentiation. However, RANKLmRNA was only detected in MALPs, but not in osteogenic cells. RANKL deficiency in MALPs induced at 3 months of age rapidly increased trabecular bone mass in long bones as well as vertebrae within 1 month due to diminished bone resorption but had no effect on the cortical bone. Ovariectomy (OVX) induced trabecular bone loss at both sites. RANKL depletion either before OVX or at 6 weeks post OVX protected and restored trabecular bone mass. Furthermore, bone healing after drill-hole injury was delayed in iCKO mice. Together, our findings demonstrate that MALPs play a dominant role in controlling trabecular bone resorption and that RANKL from MALPs is essential for trabecular bone turnover in adult bone homeostasis, postmenopausal bone loss, and injury repair.
Collapse
Affiliation(s)
| | | | - Qi He
- University of Pennsylvania
| | | | | | | | | | | | | | | | | | | | - Kai Tan
- The Children's Hospital of Philadelphia
| | - Yongwon Choi
- University of Pennsylvania Perelman School of Medicine
| | | |
Collapse
|
45
|
Zhu L, Zhang L, Cha J, Li C, Mao B. Loss of ZC4H2, an Arthrogryposis Multiplex Congenita Associated Gene, Promotes Osteoclastogenesis in Mice. Genes (Basel) 2024; 15:1134. [PMID: 39336725 PMCID: PMC11431781 DOI: 10.3390/genes15091134] [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: 08/11/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
ZC4H2 encodes a C4H2-type zinc finger protein, mutations of which lead to a spectrum of diseases known as ZC4H2 associated rare disorders (ZARD). In addition to neurological phenotypes, the most typical symptoms of ZARD are multiple joint contractures of varying degrees, accompanied by abnormal development of muscles and bones, and osteoporosis in some cases. The pathogenic mechanisms of such bone related phenotypes, however, remain unclear. Here, we showed that ZC4H2 is expressed in the developing bones in mice. ZC4H2 knockout mice were neonatal-lethal and smaller in size, with reduced calcification of long bones. Upon induced loss of ZC4H2 postnatally, the femoral bones developed an osteoporosis-like phenotype, with reduced bone mineral density, bone-volume fraction, and trabecular bone number. Knockdown of ZC4H2 showed no clear effect on the expression of osteogenic differentiation genes in in vitro models using mesenchymal stem cells. Interestingly, ZC4H2 knockdown significantly enhanced osteoclast differentiation and bone resorption in induced bone marrow-derived macrophages. We further confirmed that the number of osteoclasts in the long bone of ZC4H2 knockout mice was increased, as well as the expression of the serum bone resorption/osteoporosis marker CTX-1. Our study unveils a new role of ZC4H2 in osteoclast differentiation and bone development, providing new clues on the pathology of ZARD.
Collapse
Affiliation(s)
- Liang Zhu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (L.Z.); (J.C.); (C.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650201, China
| | - Longlong Zhang
- Academy of Biomedical Engineering, Kunming Medical University, Kunming 650500, China;
| | - Jingmei Cha
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (L.Z.); (J.C.); (C.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650201, China
| | - Chaocui Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (L.Z.); (J.C.); (C.L.)
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (L.Z.); (J.C.); (C.L.)
| |
Collapse
|
46
|
Jiang J, Ren R, Fang W, Miao J, Wen Z, Wang X, Xu J, Jin H. Lysosomal biogenesis and function in osteoclasts: a comprehensive review. Front Cell Dev Biol 2024; 12:1431566. [PMID: 39170917 PMCID: PMC11335558 DOI: 10.3389/fcell.2024.1431566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Lysosomes serve as catabolic centers and signaling hubs in cells, regulating a multitude of cellular processes such as intracellular environment homeostasis, macromolecule degradation, intracellular vesicle trafficking and autophagy. Alterations in lysosomal level and function are crucial for cellular adaptation to external stimuli, with lysosome dysfunction being implicated in the pathogenesis of numerous diseases. Osteoclasts (OCs), as multinucleated cells responsible for bone resorption and maintaining bone homeostasis, have a complex relationship with lysosomes that is not fully understood. Dysregulated function of OCs can disrupt bone homeostasis leading to the development of various bone disorders. The regulation of OC differentiation and bone resorption for the treatment of bone disease have received considerable attention in recent years, yet the role and regulation of lysosomes in OCs, as well as the potential therapeutic implications of intervening in lysosomal biologic behavior for the treatment of bone diseases, remain relatively understudied. This review aims to elucidate the mechanisms involved in lysosomal biogenesis and to discuss the functions of lysosomes in OCs, specifically in relation to differentiation, bone resorption, and autophagy. Finally, we explore the potential therapeutic implication of targeting lysosomes in the treatment of bone metabolic disorders.
Collapse
Affiliation(s)
- Junchen Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Rufeng Ren
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Weiyuan Fang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiansen Miao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zijun Wen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Haiming Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
47
|
Zhou L, Su P, Luo X, Zhong X, Liu Q, Su Y, Zeng C, Li G. Regorafenib Attenuates Osteoclasts Differentiation by Inhibiting the NF-κB, NFAT, ERK, and p38 Signaling Pathways. ACS OMEGA 2024; 9:33574-33593. [PMID: 39130575 PMCID: PMC11307286 DOI: 10.1021/acsomega.4c01308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024]
Abstract
Osteolytic diseases such as osteoporosis and neoplastic bone metastases are caused by the excessive activation of osteoclasts. Inhibiting the excessive activation of osteoclasts is a crucial strategy for treating osteolytic diseases. This study investigated the roles and mechanisms of regorafenib, a tyrosine kinase inhibitor, on osteoclasts and osteolytic diseases. We first identified the potential targets and mechanisms of regorafenib on osteoclast-related osteolytic diseases using network pharmacological analysis and molecular docking techniques. Then, we verified its role and mechanism on osteoclasts via cellular and animal experiments. Network pharmacology analysis identified 89 common targets shared by regorafenib and osteoclast-related osteolytic diseases. Enrichment analysis suggested that regorafenib may act on osteoclast-related osteolytic diseases by modulating targets such as AKT1, CASP3, MMP9, and MAPK3, regulating biological processes such as cell proliferation, apoptosis, and phosphorylation regulation, and influencing signaling pathways such as MAPK, PI3K/AKT, and osteoclast differentiation. The molecular docking results indicated that regorafenib and AKT1, CASP3, MMP9, MAPK3, and MAPK14 were stably docked. Cell experiments demonstrated that regorafenib significantly inhibited osteoclast differentiation and bone resorption in RAW 264.7 cells and bone marrow macrophages in a dose-dependent manner, with up to 50% reduction at 800 nM concentration without exhibiting cytotoxic effects. Furthermore, Western blot and RT-qPCR results demonstrated that regorafenib inhibited osteoclast differentiation by blocking the transduction of RANKL-induced NF-κB, p38, ERK, and NFAT signaling pathways. In vivo studies using an ovariectomized mouse model showed that regorafenib significantly improved bone volume fraction (BV/TV), bone surface to total volume (BS/TV), and number of trabeculae (TB.N), as well as reduced trabecular separation (Tb.Sp) compared to the OVX groups (P < 0.05). TRAcP staining results revealed a reduction in the number of osteoclasts with regorafenib treatment (P < 0.01). These results indicate that regorafenib exerts its protective effects against osteoclast-related osteolytic disease by inhibiting the RANKL-induced NF-κB, NFAT, ERK, and p38 signaling pathways. This study proves that regorafenib may serve as a potential therapeutic agent for osteoclast-related osteolytic diseases.
Collapse
Affiliation(s)
- Lin Zhou
- Department
of Endocrinology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education
Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, Guangdong, China
| | - Peiru Su
- Department
of Endocrinology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education
Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, Guangdong, China
| | - Xiangya Luo
- Department
of Endocrinology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education
Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, Guangdong, China
| | - Xuanli Zhong
- Department
of Endocrinology, The Affiliated Shunde
Hospital of Jinan University, Foshan 528305, Guangdong, China
| | - Qian Liu
- Guangxi
Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Yuangang Su
- Guangxi
Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Chunping Zeng
- Department
of Endocrinology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education
Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, Guangdong, China
| | - Ge Li
- Department
of Endocrinology, The Affiliated Shunde
Hospital of Jinan University, Foshan 528305, Guangdong, China
| |
Collapse
|
48
|
Liu Y, Chen H, Chen T, Qiu G, Han Y. The emerging role of osteoclasts in the treatment of bone metastases: rationale and recent clinical evidence. Front Oncol 2024; 14:1445025. [PMID: 39148909 PMCID: PMC11324560 DOI: 10.3389/fonc.2024.1445025] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
The occurrence of bone metastasis is a grave medical concern that substantially impacts the quality of life in patients with cancer. The precise mechanisms underlying bone metastasis remain unclear despite extensive research efforts, and efficacious therapeutic interventions are currently lacking. The ability of osteoclasts to degrade the bone matrix makes them a crucial factor in the development of bone metastasis. Osteoclasts are implicated in several aspects of bone metastasis, encompassing the formation of premetastatic microenvironment, suppression of the immune system, and reactivation of quiescent tumor cells. Contemporary clinical interventions targeting osteoclasts have proven effective in mitigating bone-related symptoms in patients with cancer. This review comprehensively analyzes the mechanistic involvement of osteoclasts in bone metastasis, delineates potential therapeutic targets associated with osteoclasts, and explores clinical evidence regarding interventions targeting osteoclasts.
Collapse
Affiliation(s)
- Youjun Liu
- Department of Spinal Surgery, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Huanshi Chen
- Department of Spinal Surgery, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Tong Chen
- Department of Spinal Surgery, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Guowen Qiu
- Department of Spinal Surgery, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Yu Han
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
49
|
Mo S, Kim MK, Jang JS, Lee SH, Hong SJ, Jung S, Kim HH. Unique expression and critical role of metallothionein 3 in the control of osteoclastogenesis and osteoporosis. Exp Mol Med 2024; 56:1791-1806. [PMID: 39085359 PMCID: PMC11372110 DOI: 10.1038/s12276-024-01290-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 08/02/2024] Open
Abstract
Bone homeostasis is maintained by an intricate balance between osteoclasts and osteoblasts, which becomes disturbed in osteoporosis. Metallothioneins (MTs) are major contributors in cellular zinc regulation. However, the role of MTs in bone cell regulation has remained unexplored. Single-cell RNA sequencing analysis discovered that, unlike the expression of other MT members, the expression of MT3 was unique to osteoclasts among various macrophage populations and was highly upregulated during osteoclast differentiation. This unique MT3 upregulation was validated experimentally and supported by ATAC sequencing data analyses. Downregulation of MT3 by gene knockdown or knockout resulted in excessive osteoclastogenesis and exacerbated bone loss in ovariectomy-induced osteoporosis. Transcriptome sequencing of MT3 knockdown osteoclasts and gene set enrichment analysis indicated that the oxidative stress and redox pathways were enriched, which was verified by MT3-dependent regulation of reactive oxygen species (ROS). In addition, MT3 deficiency increased the transcriptional activity of SP1 in a manner dependent on intracellular zinc levels. This MT3-zinc-SP1 axis was crucial for the control of osteoclasts, as zinc chelation and SP1 knockdown abrogated the promotion of SP1 activity and osteoclastogenesis by MT3 deletion. Moreover, SP1 bound to the NFATc1 promoter, and overexpression of an inactive SP1 mutant negated the effects of MT3 deletion on NFATc1 and osteoclastogenesis. In conclusion, MT3 plays a pivotal role in controlling osteoclastogenesis and bone metabolism via dual axes involving ROS and SP1. The present study demonstrated that MT3 elevation is a potential therapeutic strategy for osteolytic bone disorders, and it established for the first time that MT3 is a crucial bone mass regulator.
Collapse
Affiliation(s)
- Shenzheng Mo
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Min Kyung Kim
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Bone Science R&D Center, Tissue Regeneration Institute, Osstem Implant, Seoul, 07789, Republic of Korea
| | - Ji Sun Jang
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seung Hye Lee
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seo Jin Hong
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Suhan Jung
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea.
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
50
|
Dong S, Ge J, Meng Q, Yuan T, Wang Y, Li Y, Lu Q, Song W, Li Z, Sun S. Crebanine mitigates glucocorticoid-induced osteonecrosis of the femoral head by restoring bone remodelling homeostasis via attenuating oxidative stress. J Cell Mol Med 2024; 28:e70044. [PMID: 39205463 PMCID: PMC11358393 DOI: 10.1111/jcmm.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/24/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
The onset of osteonecrosis of the femoral head (ONFH) is intimately associated with the extensive administration of glucocorticoids (GCs). Long-term stimulation of GCs can induce oxidative stress in both osteoclasts (OCs) and osteoblasts (OBs), resulting in the disturbance of bone remodelling. An alkaloid named crebanine (CN) demonstrates pharmacological properties including anti-inflammation and reactive oxygen species (ROS) modulation. Our objective is to assess the therapeutic potential of CN in treating ONFH and elucidate the associated underlying mechanisms. The network pharmacology analysis uncovered that CN played a role in regulating ROS metabolism. In vitro, CN demonstrated its ability to reduce the dexamethasone (DEX)-stimulated generation of OCs and suppress their resorptive function by downregulating the level of osteoclast marker genes. Concurrently, CN also mitigated DEX-induced damage to OBs, facilitating the restoration of osteoblast marker gene expression, cellular differentiation and function. These effects were achieved by CN augmenting the antioxidant system to reduce intracellular ROS levels. Furthermore, in vitro results were corroborated by micro-CT and histological data, which also showed that CN attenuated MPS-induced ONFH in mice. This study highlights the therapeutic potential of CN in counteracting GCs-induced ONFH.
Collapse
Affiliation(s)
- Shankun Dong
- Department of Joint SurgeryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
| | - Jianxun Ge
- Department of Joint SurgeryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
| | - Qi Meng
- Department of Joint SurgeryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
| | - Tao Yuan
- Department of Joint SurgeryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
| | - Yi Wang
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Yi Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Qizhen Lu
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Wenao Song
- Department of Clinical LaboratoryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
| | - Ziqing Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Shui Sun
- Department of Joint SurgeryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| |
Collapse
|