1
|
Yang Y, Xiang Y, Xu P, Zhang W, Wang Y, Feng L, She R. Immuno-osteoinductive 3D printed hydrogel scaffolds with triple crosslinking and GA/EGCG release for bone healing. Colloids Surf B Biointerfaces 2025; 252:114651. [PMID: 40158247 DOI: 10.1016/j.colsurfb.2025.114651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Bone defects, caused by trauma, osteomyelitis, or osteoporosis, represent a significant global health challenge in orthopedics. However, current bone repair strategies often neglect the critical role of the immune microenvironment, which can impede effective bone regeneration. To address this gap, we developed a 3D-printed triple crosslinked hydrogel scaffold incorporating slow-release glycopyrrolate (GA) and epigallocatechin gallate (EGCG), that it could promote bone regeneration by modulating the immune response. We evaluated their immunomodulatory and bone-regenerative effects through in vitro cellular experiments and rat cranial defect models. Results demonstrated that these scaffolds effectively modulated the immune microenvironment, reducing inflammation while promoting osteoblast differentiation and proliferation, thereby significantly enhancing new bone formation and density. In conclusion, our novel 3D-printed hydrogel scaffold offers a promising approach to bone defect repair through its unique combination of mechanical strength, immunomodulation, and osteogenesis. This study provides valuable insights into leveraging immunomodulatory agents for enhanced bone regeneration, highlighting potential clinical applications.
Collapse
Affiliation(s)
- Yanlan Yang
- Department of Oral Implantation, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| | - Yang Xiang
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China.
| | - Pu Xu
- Department of Oral Implantation, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| | - Wenbo Zhang
- Department of Oral Implantation, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| | - Yawen Wang
- Department of Oral Implantation, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| | - Longbao Feng
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Rong She
- Department of Oral Implantation, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| |
Collapse
|
2
|
Wan Y, Nemoto YL, Oikawa T, Takano K, Fujiwara TK, Tsujita K, Itoh T. Mechanical control of osteoclast fusion by membrane-cortex attachment and BAR proteins. J Cell Biol 2025; 224:e202411024. [PMID: 40338171 PMCID: PMC12060795 DOI: 10.1083/jcb.202411024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 05/09/2025] Open
Abstract
Osteoclasts are multinucleated giant cells that are formed by the fusion of precursor cells. Cell-cell fusion is mediated by membrane protrusion driven by actin reorganization, but the role of membrane mechanics in this process is unknown. Utilizing live-cell imaging, optical tweezers, manipulation of membrane-to-cortex attachment (MCA), and genetic interference, we show that a decrease in plasma membrane (PM) tension is a mechanical prerequisite for osteoclast fusion. Upon RANKL-induced differentiation, ezrin expression in fusion progenitor cells is reduced, resulting in a decrease in MCA-dependent PM tension. A forced elevation of PM tension by reinforcing the MCA conversely suppresses cell-cell fusion. Mechanistically, reduced PM tension leads to membrane protrusive invadosome formation driven by membrane curvature-inducing/sensing BAR proteins, thereby promoting cell-cell fusion. These findings provide insights into the mechanism of cell-cell fusion under the control of membrane mechanics.
Collapse
Affiliation(s)
- Yumeng Wan
- Division of Membrane Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuri L. Nemoto
- Division of Membrane Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Biosignal Research Center, Kobe University, Kobe, Japan
| | - Tsukasa Oikawa
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kazunori Takano
- Department of Biology, Graduate School of Science, Chiba University, Chiba, Japan
| | - Takahiro K. Fujiwara
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
| | - Kazuya Tsujita
- Division of Membrane Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Biosignal Research Center, Kobe University, Kobe, Japan
| | - Toshiki Itoh
- Division of Membrane Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Biosignal Research Center, Kobe University, Kobe, Japan
| |
Collapse
|
3
|
Eun SY, Cheon YH, Lee CH, Chung CH, Lee MS, Kim JY. PEX5 acts as a negative regulator of RANKL-induced osteoclastogenesis in vitro and inflammatory calvarial bone destruction in vivo. Biochem Biophys Res Commun 2025; 767:151924. [PMID: 40319819 DOI: 10.1016/j.bbrc.2025.151924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Peroxisomal biogenesis factor 5 (PEX5), a peroxisomal import receptor, is well recognized for its role in protein trafficking and oxidative stress regulation. However, its function in bone metabolism remains unclear. Given the established impact of oxidative stress on osteoclast differentiation, this study explores the previously uncharacterized role of PEX5 in osteoclastogenesis and bone resorption. Using bone marrow-derived macrophages, we examined the effects of PEX5 knockdown (siPEX5) and recombinant PEX5 protein (rpPEX5) on osteoclast differentiation. Osteoclast activity was evaluated through TRAP staining, F-actin ring formation, and bone resorption assays. qRT-PCR and Western blot analyses assessed gene and protein expression, while an lipopolysaccharide (LPS)-induced calvarial bone loss model provided in vivo validation. PEX5 expression declined during osteoclast differentiation, and its suppression promoted osteoclastogenesis by increasing c-Fos, NFATc1, and osteoclast-specific gene expression. Loss of PEX5 also enhanced receptor activator of nuclear factor kappa-Β ligand (RANKL)-induced activation of Akt, MAPK, IκB, and calcium-dependent pathways, accelerating osteoclast maturation. In contrast, rpPEX5 treatment effectively inhibited osteoclast differentiation and bone resorption by downregulating NFATc1 and dampening RANKL-mediated signaling. In vivo, rpPEX5 administration mitigated LPS-induced bone loss by preserving bone structure and reducing osteoclast activity. These findings reveal a novel function of PEX5 as a regulator of osteoclast differentiation, independent of its peroxisomal role. The extracellular activity of PEX5 suggests a broader regulatory mechanism in bone metabolism, with potential therapeutic implications for osteolytic diseases.
Collapse
Affiliation(s)
- So Young Eun
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk, 54538, Republic of Korea; Department of Pharmacology, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, 54538, Republic of Korea
| | - Yoon-Hee Cheon
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk, 54538, Republic of Korea
| | - Chang Hoon Lee
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk, 54538, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan, Jeonbuk, 54538, Republic of Korea
| | - Chong Hyuk Chung
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk, 54538, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan, Jeonbuk, 54538, Republic of Korea
| | - Myeung Su Lee
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk, 54538, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan, Jeonbuk, 54538, Republic of Korea.
| | - Ju-Young Kim
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk, 54538, Republic of Korea.
| |
Collapse
|
4
|
Lee HK, Notario GR, Won SY, Kim JH, Lee SM, Kim HS, Cho SR. Elevated sclerostin levels contribute to reduced bone mineral density in non-ambulatory stroke patients. Bone Rep 2025; 25:101829. [PMID: 40225703 PMCID: PMC11986488 DOI: 10.1016/j.bonr.2025.101829] [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: 01/06/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 04/15/2025] Open
Abstract
Osteoporosis following stroke is a significant impediment to patient recovery. Decreased mechanical loading and locomotion following the onset of paralysis in stroke patients, especially those who are non-ambulatory, contributes greatly to bone loss. Sclerostin, a protein encoded by the SOST gene, accumulates as a result of reduced mechanical loading and inhibits bone formation. This study explores the relationship between mechanical unloading, sclerostin levels, and bone mineral density (BMD) in stroke patients, utilizing three cohorts. Analysis of Cohort 1, consisting of patients with available sclerostin level measurements, found significantly elevated sclerostin levels in non-ambulatory patients compared to ambulatory patients, indicating the influence of ambulatory status on sclerostin regulation. Cohort 2, consisting of patients with BMD measurements, demonstrated that prolonged mechanical unloading in non-ambulatory patients resulted in a greater decline in BMD over time. Analysis in Cohort 3 patients, who had bilateral BMD measurements available, revealed that hemiplegic sides subjected to reduced mechanical loading exhibited lower BMD compared to non-hemiplegic sides. These findings collectively confirm the hypothesis that reduced mechanical loading elevates sclerostin levels and accelerates bone loss. By integrating data across the three cohorts, this study underscores the critical impact of mechanical unloading on bone health, particularly in chronic stroke patients with limited mobility. Our study provides clinical insights for treatments integrating ambulatory status, sclerostin levels, and BMD in chronic stroke patients and highlights an increased need for therapeutics targeting mechanical loading pathways and sclerostin accumulation which can be administered to treat chronic osteoporosis following stroke.
Collapse
Affiliation(s)
- Hye Kyoung Lee
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Geneva Rose Notario
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Young Won
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Hwan Kim
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su Min Lee
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Nursing, Yonsei University College of Nursing, Seoul, Republic of Korea
| | - Ha Seong Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Seosong Hospital, Incheon, Republic of Korea
| | - Sung-Rae Cho
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
5
|
Rillaerts K, Verlinden L, Doms S, Carmeliet G, Verstuyf A. A comprehensive perspective on the role of vitamin D signaling in maintaining bone homeostasis: Lessons from animal models. J Steroid Biochem Mol Biol 2025; 250:106732. [PMID: 40122304 DOI: 10.1016/j.jsbmb.2025.106732] [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: 12/16/2024] [Revised: 02/14/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
1,25(OH)2D3 is well known for its role in maintaining normal serum calcium levels. Through its receptor, 1,25(OH)2D3 enhances intestinal calcium absorption and renal calcium reabsorption, thereby ensuring serum calcium levels are within physiological range, which is in turn important for normal bone development and mineralization. The vitamin D receptor (VDR) achieves this via transcriptional induction of genes important in calcium transport. When intestinal and renal calcium (re)absorption is impaired, VDR-mediated signaling will stimulate bone resorption and inhibit mineralization in order to maintain normal serum calcium levels, as evidenced in mice with a systemic or intestine-specific deletion of the VDR. However, VDR signaling in bone is also reported to have anabolic effects. In this review we will discuss the effects of 1,25(OH)2D3-mediated VDR signaling on bone homeostasis and provide an overview of the in vitro experiments and various transgenic mice models that have been generated to unravel the role of VDR signaling in different bone cell types such as chondrocytes, (pre)osteoblasts, osteocytes, and (pre)osteoclasts.
Collapse
Affiliation(s)
- Kayleigh Rillaerts
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| | - Lieve Verlinden
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| | - Stefanie Doms
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| | - Geert Carmeliet
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| | - Annemieke Verstuyf
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| |
Collapse
|
6
|
Woo SM, Paek K, Yoon YM, Kim H, Park SI, Kim JA. Development of a BMU-on-a-chip model based on spatiotemporal regulation of cellular interactions in the bone remodeling cycle. Mater Today Bio 2025; 32:101658. [PMID: 40206145 PMCID: PMC11979395 DOI: 10.1016/j.mtbio.2025.101658] [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: 01/06/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Bone remodeling is essential for maintaining bone homeostasis throughout life by replacing old bone with new tissue. This dynamic process occurs continuously within basic multicellular unit (BMU) through well-coordinated interactions among osteocytes, osteoblasts, and osteoclasts. However, a precise in vitro model that accurately replicates this mechanism has not yet been developed. In this study, we created a human in vitro BMU-modeling chip platform by tri-culturing cells within a chip unit integrated into a tissue culture well plate, enabling high-throughput three-dimensional (3D) cell culture. To establish the tri-culture, human osteoblasts were isolated from human surgical bone samples and differentiated into osteocytes within collagen gel inside the chip unit. Subsequently, osteoblasts and peripheral blood mononuclear cells (PBMCs) containing osteoclast precursors were added to the chip unit. To simulate each phase of the bone remodeling cycle, we optimized the tri-culture process by adjusting the timing and using two types of osteoblasts at different stages of differentiation. The completed tri-culture model successfully mimicked the bone formation phase. When receptor activators of nuclear factor kappa-Β (RANKL) and macrophage colony-stimulating factor (M-CSF) were introduced, the cells exhibited characteristics of the reversal phase, where osteogenic and osteoclastogenic environments coexist. Additionally, using more differentiated osteoblasts within the tri-culture platform induced osteoclast differentiation, resembling the bone resorption phase. Overall, our model effectively replicates each phase of the bone remodeling cycle in BMUs, both spatially and temporally. This advancement not only facilitates the study of the intricate mechanisms of bone remodeling and cellular function but also aids drug development by providing a robust bone model for testing target drugs.
Collapse
Affiliation(s)
- Sang-Mi Woo
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
| | - Kyurim Paek
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
| | - Yeo Min Yoon
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
| | - Hyang Kim
- Institute of New Horizon Regenerative Medicine, Myongji Hospital, Goyang, 10475, Republic of Korea
| | - Serk In Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, 34113, Republic of Korea
- Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, 06973, Republic of Korea
| |
Collapse
|
7
|
Wu B, Cheng Z, Li X, Liang M, Wang X, Pi D, Liu J, Li H, Zhao J, Wang J, Liang F, Wang X. The developmental toxicity of bisphenol F exposure on the zebrafish larvae. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118282. [PMID: 40344781 DOI: 10.1016/j.ecoenv.2025.118282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 05/03/2025] [Accepted: 05/04/2025] [Indexed: 05/11/2025]
Abstract
As a major substitute for the bisphenol A (BPA), the use of the bisphenol F (BPF) has increased dramatically in recent years. Growing evidence suggest that BPF shares numerous toxicological properties with BPA, raising the concerns about its potential impact on the health of organisms. However, the developmental toxicity of BPF remains poorly understood. In this study, we conducted a 5-day BPF exposure experiment on zebrafish (Danio rerio) from blastula stage at concentrations of 2, 20, 200, and 2000 µg/L. Our results demonstrated a significant increase in hatching rates across all treatment groups at 2 days post-fertilization (dpf). The esr1 was significantly upregulated at 2000 µg/L by 5 dpf, while no significant change was observed in ar. The frequency of operculum loss significantly increased at exposure concentrations of 20, 200, and 2000 µg/L, and a notable increase in notochord loss was observed at 2000 µg/L. To explore the underlying mechanisms, transcriptomic analysis was performed to identify differentially expressed genes (DEGs). GO and KEGG pathway enrichment analysis revealed that the toxic effects of BPF were closely associated with osteoclast differentiation, the FoxO signaling pathway, and the MAPK signaling pathway. These pathways influenced critical biological processes, including response to stimuli, animal organ morphogenesis, detoxification, and biomineralization. This study provides evidence that BPF exposure at environmentally relevant concentrations (2 µg/L) is harmful to hatching, concentrations above 20 µg/L exhibit estrogenic-disrupting activity and exert toxicological effects on the development of the head skeleton in zebrafish. These effects are particularly linked to disruptions in osteoclast differentiation.
Collapse
Affiliation(s)
- Biyu Wu
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Zirui Cheng
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiang Li
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Minxing Liang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xue Wang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Duan Pi
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jiayi Liu
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Huiling Li
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jun Zhao
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Junjie Wang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Fang Liang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| | - Xuegeng Wang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
8
|
Yang B, Tan M, Xiong F. Global trends in osteoimmunology and osteoporosis research: A bibliometric analysis from 2013 to 2022. Medicine (Baltimore) 2025; 104:e42367. [PMID: 40324222 PMCID: PMC12055078 DOI: 10.1097/md.0000000000042367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/28/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND A large number of studies have shown that osteoporosis is closely related to bone immunology. The purpose of this study is to conduct bibliometrics and visual analysis of the fields related to osteoimmunology and osteoporosis from 2013 to 2022 and to summarize the research hotspots and trends in this field. METHODS We searched the Web of Science core collection database for articles on osteoimmunology and osteoporosis published between 2013 and 2022. Vosviewer 1.6.18 and CiteSpace.6.2. R4 were used to analyze the retrieved data. RESULTS A total of 3218 articles on osteoimmunology and osteoporosis were included in this study. A total of 76 countries, 347 institutions, and 502 authors were included in the articles examined in this study. The main research countries were China, the United States, and South Korea. Shanghai Jiaotong University, Harvard University, and the University of California system were the main research institutions. The author who published the most papers was Xu, Jiake. CONCLUSIONS This study is the first to summarize the global research trends in the field of osteoimmunology and osteoporosis from 2013 to 2022. That helps researchers quickly understand the research hotspots and directions in this field.
Collapse
Affiliation(s)
- Bencheng Yang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Mingshuai Tan
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Fusheng Xiong
- Department of Spine Surgery, Suining Municipal Hospital of Traditional Chinese Medicine, Suining, Sichuan, China
| |
Collapse
|
9
|
Guo WY, Wu QM, Zeng HF, Chen YL, Xu J, Yu ZY, Shu YK, Yang XN, Zhang CH, He XZ, Mi JN, Chen S, Chen XM, Wu JQ, Yao HQ, Liu L, Pan HD. A sinomenine derivative alleviates bone destruction in collagen-induced arthritis mice by suppressing mitochondrial dysfunction and oxidative stress via the NRF2/HO-1/NQO1 signaling pathway. Pharmacol Res 2025; 215:107686. [PMID: 40088961 DOI: 10.1016/j.phrs.2025.107686] [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: 12/23/2024] [Revised: 02/09/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Bone destruction in rheumatoid arthritis (RA) leads to significant disability, yet effective treatments are limited. Sinomenine (Sino) demonstrates anti-arthritic and bone-protective effects but requires high doses. In this study, we developed a Sino derivative, SINX, and evaluated its efficacy in RA. Safety assessments in mice confirmed its suitability for further study. In vitro, SINX inhibited osteoclast differentiation by reducing TRAP-positive cells, disrupting F-actin ring formation, and suppressing bone resorption pits, alongside downregulating osteoclast-specific genes. It also showed strong anti-inflammatory properties by reducing inflammatory cytokine levels. In vivo, using a collagen-induced arthritis (CIA) mouse model, SINX improved bone integrity by reducing joint inflammation, maintaining trabecular bone density, and preventing erosion. Histological and micro-CT analyses confirmed its effects, including suppressed osteoclast activity and reduced bone resorption-related gene expression. Mechanistically, SINX ameliorated mitochondrial dysfunction, decreased ROS levels, and activated the NRF2/HO-1/NQO1 pathway, enhancing antioxidant defenses. Compared to Sino, SINX achieved similar results at lower doses. These findings highlight the potential of SINX as a safe, effective treatment for RA-related bone destruction.
Collapse
Affiliation(s)
- Wan-Yi Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Qi-Min Wu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Jiangning District, Nanjing 211198, China
| | - Hao-Feng Zeng
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Yu-Lian Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Jie Xu
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Zhen-Yi Yu
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Yong-Kang Shu
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Xiao-Nan Yang
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Chuan-Hai Zhang
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Xi-Zi He
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Jia-Ning Mi
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Si Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Xiao-Man Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China
| | - Jia-Qi Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - He-Quan Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Jiangning District, Nanjing 211198, China
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao
| | - Hu-Dan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome /The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province 510405, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao.
| |
Collapse
|
10
|
Huang Z, Fu Z, Wang J, Yang Z, Wang J, Yu J, Wang Z, Yang H. Effects of dietary supplementation levels of vitamin A and vitamin D 3 on growth performance, jejunal function, and tibia development in goslings from 1 to 28 days of age. Poult Sci 2025; 104:104780. [PMID: 40127567 PMCID: PMC11980002 DOI: 10.1016/j.psj.2025.104780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 03/26/2025] Open
Abstract
This study explored the interaction effects of dietary Vitamin A (VA) and Vitamin D3 (VD3) on growth performance, jejunal function, and tibia development in goslings, aiming to identify any synergistic outcomes that may reshape nutritional strategies for geese production. A total of 540 one-day-old male Jiangnan White goslings with similar body weight (82 ± 5 g) were randomly assigned into 9 treatments with five replicate pens per treatment and 12 birds per pen. The bird trial employed a 3 × 3, two-factorial treatment with three levels of VA (5000, 7000, and 9000 IU/kg) and three levels of VD3 (1000, 1500, and 2000 IU/kg) from one to 28 days of age. Main effects analysis indicated that birds fed 7000 IU/kg VA exhibited the highest ADG, BW, jejunal maltase activity and IL-10 content (P < 0.05), while 9000 IU/kg VA had the highest SOD activity and content of IL-6 and TNF-α in jejunal mucosa (P < 0.05). Both 7000 IU/kg or 9000 IU/kg VA increased the jejunal IL-1β content, relative expression of tight junction protein 1 (TJP1) mRNA, tibia defatted weight and ash weight (P < 0.05). Birds fed 2000 IU/kg VD3 exhibited the highest ADFI, while both 1500 or 2000 IU/kg VD3 increased jejunal maltase activity, and tibia ash content (P < 0.05). An interaction between VA and VD3 on ADFI, F/G, jejunal maltase activity, mucosal immune factors (IL-1β, IL-6, IL-10, TNF-α), tibia ash content, and bone morphogenetic protein-2 (BMP-2) expression. A simple effects analysis revealed that at a 5000 IU/kg VA, adding 1000 IU/kg VD3 decreased IL-1β, IL-6, TNF-α (P < 0.05). At a 7000 IU/kg VA, adding 1500 or 2000 IU/kg VD3 decreased TNF-α, and increased jejunal maltase activity(P < 0.05). At a 9000 IU/kg VA, adding 1000 IU/kg VD3 decreased ADFI, F/G, jejunal maltase activity, tibia ash, and BMP-2, while IL-1β, IL-6, and TNF-α increased (P < 0.05). At a 9000 IU/kg VA, adding 2000 IU/kg VD3 increased IL-10 (P < 0.05). At a 1000 IU/kg VD3, adding 5000 IU/kg VA increased F/G, jejunal maltase activity and IL-10, while decreased IL-1β, IL-6, TNF-α (P < 0.05), and adding 9000 IU/kg VA decreased tibia ash and BMP-2 (P < 0.05). At 1500 or 2000 IU/kg VD3, adding 7000 IU/kg VA increased jejunal maltase activity, IL-10 (P < 0.05). At a 2000 IU/kg VD3, adding 9000 IU/kg VA increased IL-6, and TNF-α (P < 0.05). In summary, a dietary level of 7000 IU/kg of VA and 2000 IU/kg of VD3 can be a balanced combination to optimize feed intake and conversion, jejunal function, and tibia mineralization, consequently enhancing growth performance in goslings.
Collapse
Affiliation(s)
- Zixin Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zhenming Fu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jian Wang
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, China
| | - Zhi Yang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jun Wang
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, China
| | - Jun Yu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, China
| | - Zhiyue Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Haiming Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
11
|
Armeni E. Navigating skeletal wellness after breast cancer. Maturitas 2025; 196:108250. [PMID: 40154015 DOI: 10.1016/j.maturitas.2025.108250] [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: 12/15/2023] [Revised: 11/07/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Breast cancer is the leading cause of death in the female population. Hormone receptor-positive cancers are usually treated with surgery in combination with endocrine therapy. The latter is known to lower estrogen levels, contributing, therefore, to loss of bone density (BMD) and higher risk of fracture. Bone-modifying agents (BMAs) can regulate the bone-related adverse effects of cancer treatment. In premenopausal women, intravenous zoledronate effectively prevents bone loss. However, the evidence regarding its ability to reduce disease recurrence remains inconclusive. In postmenopausal women, denosumab demonstrates the most substantial evidence for fracture prevention, supported by one well-powered randomized controlled trial, but has not been shown to confer anticancer benefits. While bisphosphonates effectively prevent and reduce clinical vertebra fractures, their impact on overall fracture risk is unclear. In clinical practice, management of bone health in this group of patients starts with stratification for the risk of fracture. This can be done using the FRAX algorithm; measurements of bone mineral density can help to optimize stratification for individuals at higher fracture risk. Caution is advised when interpreting the results, as the FRAX algorithm has been considered to underestimate the true fracture risk in this population, given that the algorithm has not been adjusted for the effect of anti-cancer agents. Nowadays, clodronate, ibandronate, and zoledronic acid are recommended for bone protection in this group of patients, while denosumab is not. Further research is required to highlight the optimal BMA according to patient characteristics.
Collapse
Affiliation(s)
- Eleni Armeni
- 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Athens, Greece; Royal Free Hospital NHS Trust, Medical School, UK; Department of Applied Health Sciences, School of Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
12
|
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z, Li N. Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res 2025; 71:227-262. [PMID: 38876191 DOI: 10.1016/j.jare.2024.06.010] [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: 02/17/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND As people age, degenerative bone and joint diseases (DBJDs) become more prevalent. When middle-aged and elderly people are diagnosed with one or more disorders such as osteoporosis (OP), osteoarthritis (OA), and intervertebral disc degeneration (IVDD), it often signals the onset of prolonged pain and reduced functionality. Chronic inflammation has been identified as the underlying cause of various degenerative diseases, including DBJDs. Recently, excessive activation of pyroptosis, a form of programed cell death (PCD) mediated by inflammasomes, has emerged as a primary driver of harmful chronic inflammation. Consequently, pyroptosis has become a potential target for preventing and treating DBJDs. AIM OF REVIEW This review explored the physiological and pathological roles of the pyroptosis pathway in bone and joint development and its relation to DBJDs. Meanwhile, it elaborated the molecular mechanisms of pyroptosis within individual cell types in the bone marrow and joints, as well as the interplay among different cell types in the context of DBJDs. Furthermore, this review presented the latest compelling evidence supporting the idea of regulating the pyroptosis pathway for DBJDs treatment, and discussed the potential, limitations, and challenges of various therapeutic strategies involving pyroptosis regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW In summary, an interesting identity for the unregulated pyroptosis pathway in the context of DBJDs was proposed in this review, which was undertaken as a spoiler of peaceful coexistence between cells in a degenerative environment. Over the extended course of DBJDs, pyroptosis pathway perpetuated its activity through crosstalk among pyroptosis cascades in different cell types, thus exacerbating the inflammatory environment throughout the entire bone marrow and joint degeneration environment. Correspondingly, pyroptosis regulation therapy emerged as a promising option for clinical treatment of DBJDs.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jilin Fan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000 China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300 China.
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
13
|
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
|
14
|
Mukherjee G, Samanta S, Bishayi B. TLR-4Ab and IFNγAb with exogenous IL-10 treated LPS induced mice shown differential inflammatory response upon RANKL-M-CSF stimulation in resident bone marrow cells. Microb Pathog 2025; 202:107416. [PMID: 40023455 DOI: 10.1016/j.micpath.2025.107416] [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: 10/03/2024] [Revised: 02/19/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
The inflammatory response in bone tissue often triggered by LPS is a complex process. Since LPS through TLR4 and in presence of IFNγ activates osteoclast differentiation and bone resorption, therefore, suppression of osteoclastogenesis through inhibition of TLR4 vs IFNγ mediated inflammation could be a reasonable strategy for the treatment of inflammatory bone loss. Administration of anti-TLR4 (30 mg/kg) and anti-IFNγ antibodies (6.6 mg/kg) were utilized before LPS (5 mg/kg) challenge and subsequently mice were treated with mouse IL-10 (0.02 mg/kg). Then RBMCs were isolated from different groups of mice and stimulated (in vitro) with M-CSF (10 ng/ml) and RANKL (10 ng/ml) to induce bone marrow cell differentiation in presence of LPS (100 ng/ml). The involvement of RANKL and M-CSF in the regulation of bone inflammation underlines the intricate signaling pathways. Furthermore, the study sheds light on the potential therapeutic effects of exogenous IL-10 possibly through STAT3 signaling in the RBMCs. The use of antibodies against TLR4 and IFNγ, in conjugation with IL-10in LPS bone damage model, appears to downregulate the activation of NF-κB, and reduction of many pro-inflammatory cytokines regulating the inflammatory cascade in RBMC. This suggests a promising avenue for the development of treatments aimed at mitigating bone inflammation associated with bacterial infections. Therefore, inhibition of TLR4 and IFNγ could be explored as potential therapeutic agents against LPS induced bone loss.
Collapse
Affiliation(s)
- Gopinath Mukherjee
- Department of Physiology, Immunology and Microbiology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, 700009, West Bengal, India
| | - Sharmistha Samanta
- Department of Physiology, Immunology and Microbiology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, 700009, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology and Microbiology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, 700009, West Bengal, India.
| |
Collapse
|
15
|
Daljeet M, Warunek S, Covell DA, Monegro A, Giangreco T, Al-Jewair T. Association between obstructive sleep apnea syndrome and bone mineral density in adult orthodontic populations. Cranio 2025; 43:390-400. [PMID: 36368042 DOI: 10.1080/08869634.2022.2142724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To determine the association between obstructive sleep apnea syndrome (OSAS) and predicted bone mineral density (BMD) in adults presenting for orthodontic treatment. METHODS This retrospective cross-sectional study included 38 adults divided into OSAS and non-OSAS groups. Using pre-treatment CBCT images, radiographic density (RD) of left and right lateral regions of the 1st cervical vertebrae and dens of the 2nd cervical vertebrae were measured as an indicator for BMD. RESULTS When controlling for age, sex, and BMI, the mean RD was significantly lower in the OSAS group compared to the non-OSAS group (left CV1: 36.69 ± 84.50 vs. 81.67 ± 93.25 Hounsfield Units [HU], respectively, p = 0.031; right CV1: 30.59 ± 81.18 vs. 74.26 ± 91.81 HU, p = 0.045; dens: 159.25 ± 115.96 vs. 223.94 ± 106.09 HU, p = 0.038). CONCLUSION Adults with OSAS have lower values for predicted BMD than those without OSAS.
Collapse
Affiliation(s)
| | - Stephen Warunek
- Department of Orthodontics, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - David A Covell
- Department of Orthodontics, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - Alberto Monegro
- Pediatric Sleep Center, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | | | - Thikriat Al-Jewair
- Department of Orthodontics, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
16
|
Liu Y, Zhu Y, Zhao Y, Wang J, Yu Y, Zhu J, Jin G. Bovine collagen peptides and peptide-calcium complexes inhibit RANKL-induced osteoclast differentiation in RAW 264.7 macrophages. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025. [PMID: 40312763 DOI: 10.1002/jsfa.14308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/09/2025] [Accepted: 03/03/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND This study aims to prepare collagen peptides from bovine bone meal using a combination of heat pretreatment and enzymatic digestion, and to chelate them with calcium chloride to form peptide-calcium chelates. The effects of both on the proliferation and differentiation of osteoclasts were investigated using cellular experiments (RAW 264.7 cells). RESULTS Both bovine collagen peptides and their calcium chelates (BPs, HBPs, BPs-Ca, and HBPs-Ca) can significantly inhibit the RANKL-induced differentiation of RAW 264.7 cells into osteoclasts. The preheating treatment before enzymatic hydrolysis of bone materials has an improving effect on the inhibition of RAW 264.7 differentiation into osteoblasts by collagen peptides and their peptide calcium chelates. HBP and HBPs-Ca could significantly activate the NF-κB signaling pathway, among which HBPs-Ca was the most effective, which could significantly downregulate the mRNA expression of genes related to osteoclast differentiation, such as AP-1, c-Fos, TRAP, and NFATc1. Additionally, the expression of NF-κB p65, c-Fos, IKK and IκBα were also significantly inhibited after treatment with HBPs-Ca, with IKK being the most significantly downregulated, with an 8.2-fold reduction compared to the control group. CONCLUSION HBPs and HBPs-Ca demonstrated stronger activity in inhibiting osteoclast formation compared to BPs and BPs-Ca. This enhanced activity is likely due to structural changes in the peptides caused by heat treatment, which increase their antioxidant properties and antagonistic effects on RANKL. These findings indicate that bovine collagen peptides and their calcium chelates can inhibit the formation of osteoclasts by activating the NF-κB pathway, thereby influencing bone metabolism and providing a theoretical basis for the treatment of osteoporosis. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yuanyi Liu
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, School of Food and Health, Beijing of Technology and Business University, Beijing, 100048, China
| | - Yue Zhu
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, School of Food and Health, Beijing of Technology and Business University, Beijing, 100048, China
| | - Yixin Zhao
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, School of Food and Health, Beijing of Technology and Business University, Beijing, 100048, China
| | - Jin Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, School of Food and Health, Beijing of Technology and Business University, Beijing, 100048, China
| | - Yiying Yu
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, School of Food and Health, Beijing of Technology and Business University, Beijing, 100048, China
| | - Jin Zhu
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, School of Food and Health, Beijing of Technology and Business University, Beijing, 100048, China
| | - Guofeng Jin
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, School of Food and Health, Beijing of Technology and Business University, Beijing, 100048, China
| |
Collapse
|
17
|
Kurihara T, Shimamura M, Etani Y, Noguchi T, Fukuda Y, Ochiai N, Goshima A, Miura T, Hirao M, Sugimoto A, Ju N, Yamakawa S, Kanamoto T, Nakata K, Okada S, Ebina K. RANKL-derived peptide MHP1-AcN attenuates ovariectomy-induced osteoporosis by targeting RANK and TNFR1 in mice. Bone 2025; 194:117440. [PMID: 40032017 DOI: 10.1016/j.bone.2025.117440] [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: 12/30/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
PURPOSE Estrogen deficiency following menopause increases receptor activator of nuclear factor-kappa B ligand (RANKL) expression in osteoblasts, thereby promoting osteoclast differentiation, and enhances T cell-derived tumor necrosis factor-alpha (TNFα) production, which induces sclerostin expression in osteocytes, thereby inhibiting bone formation. This study aimed to develop a novel uncoupling therapeutic agent for osteoporosis. METHODS We developed microglial healing peptide 1 with N-terminal acetylation and C-terminal amidation (MHP1-AcN), a modified RANKL peptide with N-terminal acetylation and C-terminal amidation lacking the osteoclast activating CD loop. Given the structural similarities of RANK and TNF receptor 1 (TNFR1), we hypothesized that MHP1-AcN could inhibit both the RANKL-RANK and TNFα-TNFR1 pathways to address the pathophysiology of osteoporosis, as evaluated in vitro and in vivo using an ovariectomized mouse model. RESULTS In ovariectomized mice, MHP1-AcN inhibited osteoclastogenesis, reduced osteocytic sclerostin expression, prevented bone loss, and improved the femoral cancellous and cortical bone microarchitecture. Unlike anti-RANKL antibody, MHP1-AcN considerably preserved bone formation by osteoblasts and enhanced bone strength, as evidenced by increases in energy absorption capacity. In vitro, MHP1-AcN bound to both RANK and TNFR1, suppressing osteoclast activity via the RANKL-RANK pathway and reducing sclerostin expression through the TNFα-TNFR1-nuclear factor-kappa B pathway. MHP1-AcN did not affect osteoblast proliferation and differentiation or RANKL expression. CONCLUSION MHP1-AcN effectively inhibits osteoclastogenesis and sclerostin-mediated suppression of bone formation while considerably preserving osteoblast function. These findings suggest that MHP1-AcN, which targets dual pathways critical for bone homeostasis, is a promising uncoupling therapeutic agent for osteoporosis.
Collapse
Affiliation(s)
- Takuya Kurihara
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Munehisa Shimamura
- Department of Gene & Stem Cell Regenerative Therapy, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yuki Etani
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Sports Medical Biomechanics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takaaki Noguchi
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yuji Fukuda
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Nagahiro Ochiai
- Department of Musculoskeletal Regenerative Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Atsushi Goshima
- Department of Orthopaedic Surgery, Osaka Rosai Hospital, 1179-3 Nagasonecho, Kita-ku, Sakai, Osaka 591-8025, Japan
| | - Taihei Miura
- Clinical and Research Institute for Foot and Ankle Surgery, Jujo Hospital, 341-1 Mangoku, Kisarazu, Chiba 292-0003, Japan
| | - Makoto Hirao
- Department of Orthopaedic Surgery, National Hospital Organization Osaka Minami Medical Center, 2-1 Kidohigashi-machi, Kawachinagano, Osaka 586-8521, Japan
| | - Atsushi Sugimoto
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Nan Ju
- Department of Gene & Stem Cell Regenerative Therapy, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Satoshi Yamakawa
- Department of Medicine for Sports and Performing Arts, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takashi Kanamoto
- Department of Medicine for Sports and Performing Arts, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ken Nakata
- Department of Medicine for Sports and Performing Arts, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Seiji Okada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kosuke Ebina
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Sports Medical Biomechanics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
18
|
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
|
19
|
Zhang Y, Yu T, Xiang Q, van den Tillaart F, Ma J, Zhuang Z, Stessuk T, Wang H, van den Beucken JJJP. Osteoclasts drive bone formation in ectopic and orthotopic environments. Biomaterials 2025; 322:123377. [PMID: 40319679 DOI: 10.1016/j.biomaterials.2025.123377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
To date, cell-based approaches to stimulate bone formation have primarily focused on mesenchymal stromal cells (MSCs) for their supposed osteogenic potential, but despite some pre-clinical successes, clinical outcomes have remained unsatisfactory. Emerging data suggest that osteoclasts play crucial roles in stimulating bone formation beyond their catabolic function in bone resorption. Interestingly, osteoclastic activity precedes osteoblastic bone formation in the physiological bone remodeling cycle. To explore the role of osteoclasts in bone formation further, we prepared osteoclast-based constructs and implanted them (i) ectopically to evaluate their potential to induce bone formation, and (ii) orthotopically to evaluate effects on bone regeneration. Remarkably, constructs containing primary mouse osteoclasts showed consistent and robust de novo bone formation, which presented comparable osteogenic efficacy to BMP-2 treatment. Additionally, we observed de novo bone marrow formation upon ectopic implantation of osteoclast-based constructs (incidence 73 %) and BMP-2 loaded controls (incidence 91 %). Importantly, constructs containing macrophages (MФs) or scaffold only (negative control) showed neither bone nor bone marrow formation. Further, a mouse cranial defect model confirmed the stimulatory bone regeneration capabilities of Osteoclast-based constructs, evidenced by 2.5-fold increased bone formation compared to scaffold only. These findings demonstrate the osteoinduction and osteogenesis capacity of osteoclasts, reshaping our understanding of their role in bone formation and opening new avenues for the design and development of cell-based constructs for bone repair.
Collapse
Affiliation(s)
- Yang Zhang
- School of Dentistry, Shenzhen University Medical School, Shenzhen, 518055, China; Regenerative Biomaterials, Department of Dentistry, Radboudumc, Nijmegen, 6525GA, the Netherlands
| | - Taozhao Yu
- School of Dentistry, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Qianfeng Xiang
- Regenerative Biomaterials, Department of Dentistry, Radboudumc, Nijmegen, 6525GA, the Netherlands
| | - Femke van den Tillaart
- Regenerative Biomaterials, Department of Dentistry, Radboudumc, Nijmegen, 6525GA, the Netherlands
| | - Jinling Ma
- School of Stomatology, Capital Medical University, Beijing, 100029, China
| | - Zhumei Zhuang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, 116024, China
| | - Talita Stessuk
- Regenerative Biomaterials, Department of Dentistry, Radboudumc, Nijmegen, 6525GA, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5612AZ, the Netherlands
| | - Huanan Wang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, 116024, China.
| | | |
Collapse
|
20
|
Yue Y, Cao S, Cao F, Wei Y, Li A, Wang D, Liu P, Zeng H, Lin J. Unveiling research hotspots: a bibliometric study on macrophages in musculoskeletal diseases. Front Immunol 2025; 16:1519321. [PMID: 40356917 PMCID: PMC12066445 DOI: 10.3389/fimmu.2025.1519321] [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: 10/29/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
Research on the role of macrophages in musculoskeletal (MSK) diseases has significantly increased in recent years. However, a thorough evaluation of the developmental trajectory of this field, including the contributions of prominent authors and primary research themes, remains insufficient. Furthermore, the identification of emerging research hotspots requires more detailed exploration. This study collated articles and reviews addressing "macrophages in MSK diseases" published between 2004 and 2023, with all data extracted from the Web of Science database. The collected data were analyzed using a variety of bibliometric and visualization tools, such as VOSviewer, CiteSpace, GraphPad Prism, and R packages. Results indicate that China and the United States are the leading contributors in this research domain. Among the many academic institutions involved, Shanghai Jiao Tong University and the University of California stand out as the most productive. The journal "Frontiers in Immunology" had the highest publication output on this topic. The five most frequently explored research domains include Immunology, Rheumatology, Pharmacology and Pharmacy, Cell Biology, and Biochemistry and Molecular Biology. These results offer a comprehensive overview of the current state of research in this field and provide meaningful insights for guiding future studies.
Collapse
Affiliation(s)
- Yaohang Yue
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
- Shandong Second Medical University, Clinical Medical College, Weifang, China
| | - Siyang Cao
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fuyang Cao
- Department of Orthopedics, Second Hospital of Shanxi Medical University,
Taiyuan, Shanxi, China
| | - Yihao Wei
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic
University, Hong Kong, Hong Kong SAR, China
| | - Aikang Li
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
| | - Deli Wang
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peng Liu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Zeng
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jianjing Lin
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
21
|
Guo Z, Huang Y, Wang X, Han Y, Li A, Qu Y, Chen L, Du M, Zhang Y, Xu Y. Ergothioneine alleviates osteoporosis via the ROS-MAPK signaling Axis. Bone 2025; 197:117496. [PMID: 40287031 DOI: 10.1016/j.bone.2025.117496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
The accumulation of reactive oxygen species (ROS) within cells regulates the formation and function of osteoclasts, which is crucial therapeutic target for the treatment of osteoporosis. Ergothioneine (EGT) is a rare amino acid with strong antioxidant and anti-inflammatory properties. However, its application on osteoporosis has not been reported. In this study, we investigated the effects of EGT on osteoclastogenesis in vitro and in ovariectomized (OVX) mice. The results revealed that EGT could suppress RANKL-induced podosome belt formation and osteoclast development in vitro, while reducing intracellular ROS levels by upregulating key antioxidant enzymes, including HO-1 and Catalase. EGT was also found to downregulate the expression of critical osteoclast-specific proteins such as Trap, c-Fos, and Ctsk through attenuation of MAPK signaling. The potential of EGT to protect against trabecular bone loss in OVX mice was further demonstrated by micro-CT imaging, possibly by reducing osteoclast numbers shown by histological outcomes. These findings together highlighted the potential value of EGT as a novel tool for treating osteoporosis through its ability to suppress osteoclastogenesis and mitigate the accumulation of intracellular ROS.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200072 Shanghai, PR China; School of Clinical Medicine, Jinggangshan University, 343009 Ji'an, Jiangxi, China
| | - Yiwen Huang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200072 Shanghai, PR China
| | - Xiaowei Wang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200072 Shanghai, PR China.
| | - Yi Han
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200072 Shanghai, PR China.
| | - Ang Li
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200072 Shanghai, PR China; Medical College, Anhui University of Science and Technology, Huainan, China
| | - Yiyang Qu
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200072 Shanghai, PR China
| | - Lin Chen
- Department of Gastrointestinal Surgery, Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Meihang Du
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200072 Shanghai, PR China.
| | - Yiming Zhang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200072 Shanghai, PR China.
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200072 Shanghai, PR China.
| |
Collapse
|
22
|
Ke D, Gao T, Dai H, Xu J, Ke T. RANKL promotes MT2 degradation and ROS production in osteoclast precursors through Beclin1-dependent autophagy. Differentiation 2025; 143:100863. [PMID: 40267773 DOI: 10.1016/j.diff.2025.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
ROS produced under oxidative stress are crucial for osteoclast differentiation. Metallothionein (MT) is a ROS-scavenging molecule. As a member of MT family, MT2 can clear ROS in osteoclast precursors (OCPs) and contributes to osteoclast differentiation. RANKL can promote OCP autophagy. Given the molecular-degrading effect of autophagy, the relationship between RANKL-dependent autophagy, MT2 and ROS during osteoclast differentiation is worth exploring. We depended in vitro RANKL administration and RANKL-overexpressing (Tg-RANKL) mice to observe the effects of RANKL on ROS production, MT2 protein expression, Beclin1 expression and autophagic activity in OCPs. Spautin1 was used to investigate the relationship between Beclin1-dependent autophagy and RANKL-regulated MT2 expression. Osteoclast-targeting MT2-cDNA-AAVs were applied to assess the therapeutic effect of MT2 on Tg-RANKL-related bone loss. The results showed that RANKL promoted ROS production but reduced MT2 protein expression in OCPs. RANKL also enhanced Beclin1 expression and LC3-puncta abundance. Decreased Beclin1 expression with spautin1 blocked RANKL-increased ROS production and osteoclast differentiation and recovered RANKL-decreased MT2 expression. MT2 selective overexpression with CD11b-promoter-MT2-cDNA-AAVs attenuated ROS production and osteoclastogenesis in Tg-RANKL mice and improved bone loss. Overall, RANKL can reduce MT2 protein expression through Beclin1-dependent autophagy, thereby promoting ROS production and osteoclast differentiation; this suggests that MT2-overexpressing small molecule drugs have the potential to treat RANKL-related bone loss.
Collapse
Affiliation(s)
- Dianshan Ke
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China
| | - Tingwei Gao
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China
| | - Hanhao Dai
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China
| | - Jie Xu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China.
| | - Tie Ke
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China.
| |
Collapse
|
23
|
Kim K, Kim JH, Kim I, Seong S, Kook H, Koh JT, Kim N. Tripartite motif-containing 27 negatively regulates NF-κB activation in bone remodeling. Mol Med 2025; 31:141. [PMID: 40251491 PMCID: PMC12008848 DOI: 10.1186/s10020-025-01204-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Tripartite motif-containing 27 (TRIM27) is highly expressed in the mouse thymus, spleen, and hematopoietic compartment cells and regulates cell proliferation, apoptosis, and innate immune responses. However, the role of TRIM27 in bone remodeling remains unknown. This study aimed to investigate the role of TRIM27 in the differentiation of osteoclasts and osteoblasts. METHODS We measured the effects of overexpression or knockdown of TRIM27 in osteoclasts and osteoblasts using real-time PCR and Western blot analysis to quantify the mRNA and protein levels of marker genes. Additionally, we performed an in vivo analysis of TRIM27 knockout mice through bone mineral density analysis and histological analysis. RESULTS TRIM27 deficiency decreased bone mineral density by enhancing osteoclast differentiation and inhibiting osteoblast differentiation. Overexpression of TRIM27 in osteoclast precursors suppressed osteoclast formation and resorption activity, and ectopic expression of TRIM27 in osteoblast precursors induced osteoblast differentiation and mineralization. Additionally, we found that TRIM27 attenuated NF-κB activation in both osteoclasts and osteoblasts by interacting with TAB2 and promoting TAB2 degradation through lysosomal-dependent pathways, thereby inhibiting NF-κB signaling. CONCLUSIONS Our results identify TRIM27 as a novel negative regulator of NF-κB in bone remodeling, suggesting that regulating TRIM27 may be useful in developing treatments for musculoskeletal diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
24
|
Zhang M, Xiong W, Qiao R, Li M, Zhang C, Yang C, Zhu Y, He J, Ma Z. Irisin in the modulation of bone and cartilage homeostasis: a review on osteoarthritis relief potential. Front Physiol 2025; 16:1570157. [PMID: 40313878 PMCID: PMC12043700 DOI: 10.3389/fphys.2025.1570157] [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: 02/18/2025] [Accepted: 04/04/2025] [Indexed: 05/03/2025] Open
Abstract
Osteoarthritis, a progressive and degenerative joint disease, disrupts the integrity of the entire joint structure, underscoring the urgency of identifying more effective therapeutic strategies and innovative targets. Among these, exercise therapy is considered a key component in the early management of osteoarthritis, functioning by stimulating the secretion of myokines from the skeletal muscle system. Irisin, a myokine predominantly secreted by skeletal muscle during exercise and encoded by the FNDC5 gene, has garnered attention for its regulatory effects on bone health. Emerging evidence suggests that irisin may play a protective role in osteoarthritis by promoting tissue homeostasis, enhancing subchondral bone density and microstructure, and inhibiting chondrocyte apoptosis. By improving chondrocyte viability, preserving extracellular matrix integrity, and maintaining homeostasis in osteoblasts, osteoclasts, and osteocytes, irisin emerges as a promising therapeutic target for osteoarthritis. This review delves into the role of irisin in osteoarthritis pathogenesis, highlighting its influence on cartilage and bone metabolism as well as its dynamic relationship with exercise. Additionally, this review suggests that further exploration on its specific molecular mechanisms, optimization of drug delivery systems, and strategic utilization of exercise-induced benefits will be pivotal in unlocking the full potential of irisin as a novel intervention for osteoarthritis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yan Zhu
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiaying He
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zhigui Ma
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
25
|
Shi M, Feng J, Chen P, Zhu B, Sun L, Ma Y, Zhang Y, Wang X. Targeted Dual Microdroplets for Modulating Osteoclast Differentiation and Function: A Novel Therapeutic Approach to Combat Osteoporosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22232-22244. [PMID: 40181685 PMCID: PMC12012778 DOI: 10.1021/acsami.4c21489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Osteoporosis, a condition marked by reduced bone mass and structural deterioration, continues to be a major public health concern, especially as global populations age. Excessive osteoclast formation is a hallmark of osteoporosis. The transcription factor nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) is indispensable for the early differentiation of osteoclasts, orchestrating the expression of essential genes, while at the later stages, cathepsin K (CTSK) is essential for bone resorption activities of mature osteoclasts. Here, we fabricated ultrasound-responsive microdroplets (MDs) by modulating both the early stages of osteoclast differentiation and the functions of mature osteoclasts via targeting the NFATc1 and CTSK. The internalization of these dual MDs was evaluated in human bone marrow-derived mesenchymal stromal cells (hBMSCs) and murine RAW 264.7 macrophages, alongside the biocompatibility assay. Their effects on osteogenesis and osteoclastogenesis were further investigated in vitro, followed by in vivo analysis in osteoporotic rat models. The dual MDs exhibited a well-defined core-shell structure and demonstrated efficient cellular uptake with minimal cytotoxicity. Furthermore, dual MDs showed a minimal effect on the osteogenic differentiation of the hBMSCs. In in vitro osteoclastogenesis assays, dual MDs effectively suppressed both osteoclast differentiation and formation through a synergistic inhibitory effect. In vivo studies demonstrated that osteoporotic rats receiving dual MDs showed significant protection against bone loss induced by ovariectomy. These results highlight the potential of dual MDs as a sophisticated, targeted therapeutic approach to osteoporosis treatment.
Collapse
Affiliation(s)
- Maobiao Shi
- Department
of Orthopaedic Surgery, Affiliated Hospital
of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Jin Feng
- Department
of Clinical Laboratory, Affiliated Hospital
of Zunyi Medical University, Zunyi, Guizhou 563099, China
| | - Ping Chen
- Department
of Orthopaedic Surgery, Affiliated Hospital
of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Binsong Zhu
- Department
of Orthopaedic Surgery, Affiliated Hospital
of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Langlang Sun
- Department
of Orthopaedic Surgery, Affiliated Hospital
of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yaping Ma
- Department
of Orthopaedic Surgery, Affiliated Hospital
of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yi Zhang
- Department
of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, China
- Key Laboratory
of Maternal and Child Health and Exposure Science, Guizhou Provincial
Department of Education, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Xin Wang
- Department
of Orthopaedic Surgery, Affiliated Hospital
of Zunyi Medical University, Zunyi, Guizhou 563003, China
- Guizhou Provincial
Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, Guizhou 563000, China
| |
Collapse
|
26
|
Liu C, Zuo M, Zhao J, Niu T, Hu A, Wang H, Zeng X. DPHB inhibits osteoclastogenesis by suppressing NF-κB and MAPK signaling and alleviates inflammatory bone destruction. Int Immunopharmacol 2025; 152:114377. [PMID: 40043357 DOI: 10.1016/j.intimp.2025.114377] [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: 11/09/2024] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025]
Abstract
Overactivation of osteoclasts disrupt the delicate balance between bone-resorbing osteoclasts and bone-forming osteoblasts, resulting in development of osteoporosis and various inflammatory disorders, including rheumatoid arthritis, spondyloarthropathies, and psoriatic arthritis. While inhibiting osteoclastogenesis represents a promising therapeutic strategy, current treatments targeting the RANKL pathway (such as bisphosphonates and denosumab) are associated with severe side effects, necessitating the development of safer alternatives. We hypothesize that the compound (4E)-7-(4-hydroxy-3-methoxyphenyl)-1-phenylhept-4-en-3-one (DPHB), derived from Alpinia officinarum Hance, can effectively inhibit RANKL-induced osteoclastic activity while maintaining a favorable safety profile. Through tartrate-resistant acid phosphatase (TRAP) staining and bone resorption pit assays, we demonstrate that DPHB inhibits osteoclast formation and function. Mechanistically, DPHB suppresses both NF-κB and MAPK signaling pathways while inhibiting NFATc1 activation and nuclear translocation, as evidenced by immunofluorescence staining, real-time PCR, and western blotting assays. In a LPS-induced inflammatory osteolysis mouse model, intraperitoneal administration of DPHB significantly alleviated bone destruction, confirmed through Micro-CT scanning, histological staining, and enzyme-linked immunosorbent assay (ELISA). Our findings establish DPHB as a promising osteoclast inhibitor for treating bone loss disorders through its dual suppression of osteoclastogenesis via NF-κB and MAPK signaling pathways and amelioration of inflammatory bone destruction.
Collapse
Affiliation(s)
- Caixia Liu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199, Hainan, PR China
| | - Min Zuo
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Department of Spine Surgery, Hainan Province Clinical Medical Center, The First Affiliated Hospital,Hainan Medical University, Haikou 571199, Hainan, PR China
| | - Jing Zhao
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Department of Spine Surgery, Hainan Province Clinical Medical Center, The First Affiliated Hospital,Hainan Medical University, Haikou 571199, Hainan, PR China
| | - Tianqi Niu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199, Hainan, PR China
| | - Aihua Hu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199, Hainan, PR China
| | - Hua Wang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Department of Spine Surgery, Hainan Province Clinical Medical Center, The First Affiliated Hospital,Hainan Medical University, Haikou 571199, Hainan, PR China.
| | - Xiangzhou Zeng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199, Hainan, PR China.
| |
Collapse
|
27
|
Schröder A, Fischer F, Reinert B, Jantsch J, Proff P, Paddenberg-Schubert E, Kirschneck C. During high salt treatment myeloid p38α/MAPK fosters osteoclast activity and inflammatory macrophage responses promoting orthodontic tooth movement. Front Immunol 2025; 16:1571268. [PMID: 40303394 PMCID: PMC12038906 DOI: 10.3389/fimmu.2025.1571268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/19/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction During orthodontic tooth movement, sterile inflammatory processes and alveolar bone resorption occur in the periodontal ligament, involving myeloid cells such as macrophages and osteoclasts. The myeloid p38α/MAPK (mitogen-activated protein kinase) not only regulates the inflammatory response of macrophages and osteoclast differentiation but also the activation of the osmoprotective transcription factor NFAT5 (nuclear factor of activated T cells 5) under high-salt conditions. Therefore, this study aims to investigate the relative role of myeloid p38α/MAPK in orthodontic tooth movement as a function of extracellular salt content. Material and methods Macrophages and osteoclasts were differentiated from the bone marrow of mice lacking p38α/MAPK expression in myeloid cells (p38α Δmyel) and controls for RNA analysis and calcium phosphate resorption assay. Controls and p38α Δmyel mice were fed a low or a high salt diet for a total of two weeks. One week after the start of the diet, an elastic band was inserted between the first and second molar to induce orthodontic tooth movement. Atomic absorption spectrometry was used to assess the sodium balance of the jaw bone tissue. RNA was isolated from the periodontium of the first molar, osteoclast numbers and extent of orthodontic tooth movement were assessed. Results Nfat5 mRNA was increased in macrophages and osteoclasts in vitro and in the periodontium in vivo after high salt treatment in control mice but not in p38α Δmyel mice. While there was no salt effect on interleukin-6 (Il6) gene expression, prostaglandin endoperoxide synthase-2 (Ptgs2) mRNA was upregulated in control but not in p38α Δmyel mice in vitro and in vivo. p38α/MAPK deletion increased osteoclast numbers after low and high salt diet. Of note, deletion of p38α/MAPK elevated osteoclast activity under control salt conditions but reduced osteoclast activity under high salt conditions. High-salt diet resulted in increased sodium ion deposition in the jaw of both genotypes, while tooth movement was only increased in control mice. In p38α Δmyel mice, high salt diet reduced the extent of orthodontic tooth movement, which could be explained by the reduced bone resorption of osteoclasts. Conclusion We conclude that myeloid p38α/MAPK promotes macrophage Ptgs2 expression and osteoclast activity in response to extracellular salt levels, thereby supporting orthodontic tooth movement.
Collapse
Affiliation(s)
- Agnes Schröder
- Department of Orthodontics, University Hospital Regensburg, Regensburg, Germany
- Institute for Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
| | - Florian Fischer
- Department of Orthodontics, University Hospital Regensburg, Regensburg, Germany
| | - Beatrice Reinert
- Department of Orthodontics, University Hospital Regensburg, Regensburg, Germany
| | - Jonathan Jantsch
- Institute for Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Peter Proff
- Department of Orthodontics, University Hospital Regensburg, Regensburg, Germany
| | | | | |
Collapse
|
28
|
Chandra A, Law SF, Pignolo RJ. Changing landscape of hematopoietic and mesenchymal cells and their interactions during aging and in age-related skeletal pathologies. Mech Ageing Dev 2025; 225:112059. [PMID: 40220914 DOI: 10.1016/j.mad.2025.112059] [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: 12/17/2024] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Aging profoundly impacts mesenchymal and hematopoietic lineage cells, including their progenitors-the skeletal stem cells (SSCs) and hematopoietic stem cells (HSCs), respectively. SSCs are crucial for skeletal development, homeostasis, and regeneration, maintaining bone integrity by differentiating into osteoblasts, adipocytes, and other lineages that contribute to the bone marrow (BM) microenvironment. Meanwhile, HSCs sustain hematopoiesis and immune function. With aging, SSCs and HSCs undergo significant functional decline, partly driven by cellular senescence-a hallmark of aging characterized by irreversible growth arrest, secretion of pro-inflammatory factors (senescence associated secretory phenotype, SASP), and impaired regenerative potential. In SSCs, senescence skews lineage commitment toward adipogenesis at the expense of osteogenesis, contributing to increased bone marrow adiposity (BMAd), reduced bone quality, and osteoporosis. Similarly, aged HSCs exhibit diminished self-renewal, biased differentiation, and heightened inflammation, compromising hematopoietic output and immune function. In this review, we examine the age-related cellular and molecular changes in SSCs and HSCs, their lineage decisions in the aging microenvironment, and the interplay between skeletal and hematopoietic compartments. We also discuss the role of senescence-driven alterations in BM homeostasis and how targeting cellular aging mechanisms may offer therapeutic strategies for mitigating age-related skeletal and hematopoietic decline.
Collapse
Affiliation(s)
- Abhishek Chandra
- Department of Physiology and Biomedical Engineering; Department of Medicine, Divisions of Hospital Internal Medicine and Section on Geriatric Medicine and Gerontology; Robert and Arlene Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA.
| | - Susan F Law
- Robert and Arlene Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert J Pignolo
- Department of Physiology and Biomedical Engineering; Department of Medicine, Divisions of Hospital Internal Medicine and Section on Geriatric Medicine and Gerontology; Robert and Arlene Kogod Aging Center, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
29
|
Barbanente A, Di Cosola AM, Niso M, D'Anna L, Rubino S, Indelicato S, Pacifico C, Terenzi A, Margiotta N. New oxaliplatin-zoledronate derivatives with potential antitumor activity towards bone tumors and metastases. J Inorg Biochem 2025; 270:112916. [PMID: 40245639 DOI: 10.1016/j.jinorgbio.2025.112916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 04/19/2025]
Abstract
Bone cancer can originate from any cellular element of the bone and may occur sporadically or as a result of degeneration from a precursor lesion. Bone metastases, often stemming from primary cancers such as breast and prostate cancer, are extremely painful and challenging to treat. The treatment of primary bone malignancies typically involves surgery, radiotherapy, chemotherapy and analgesics. Platinum (Pt)-based drugs are effective against bone cancers and metastases but are often limited by severe side effects due to poor specificity. To improve drug targeting and reduce systemic toxicity, bisphosphonate (BP) ligands, which selectively accumulate in bone tissue, have been combined with Pt-based drugs. The combination of Pt drugs with bisphosphonates like zoledronic acid (ZL) could lead to enhanced therapeutic outcomes due to its intrinsic pharmacological activity. In this study, we report the synthesis and full characterization of two new dinuclear Pt(II) complexes that combine ZL with clinically approved oxaliplatin and the drug-candidate kiteplatin, respectively. These complexes were tested for their stability under physiological and acidic conditions, reactivity with 5'-GMP interaction with B- and G-quadruplex DNA models and cytotoxicity against a panel of human tumor cell lines.
Collapse
Affiliation(s)
- Alessandra Barbanente
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Anna Maria Di Cosola
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Mauro Niso
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy.
| | - Luisa D'Anna
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy
| | - Simona Rubino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy
| | - Sergio Indelicato
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Chromatography and Mass Spectrometry Section, Quality Control and Chemical Risk (CQRC), Via Antonino Cassarà, 4, Palermo 90146, Italy
| | - Concetta Pacifico
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Alessio Terenzi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy.
| | - Nicola Margiotta
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
30
|
Lu Z, Xiao P, Liu S, Huang C, Li W, Mao Y, Xu Y, Tian Y. Osteoimmunology: Crosstalk Between T Cells and Osteoclasts in Osteoporosis. Clin Rev Allergy Immunol 2025; 68:41. [PMID: 40208457 DOI: 10.1007/s12016-025-09046-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2025] [Indexed: 04/11/2025]
Abstract
Osteoporosis, a common metabolic condition that affects the bones, increases the risk of fractures, thereby diminishing one's quality of life and, in severe cases, can even result in life-threatening conditions. Osteoporosis is becoming increasingly prevalent worldwide as the population ages. Previous research on osteoporosis has focused on skeletal cellular components such as osteoblasts and osteoclasts. The emerging field of "osteoimmunology" has recently been introduced through new research. The concept highlights the critical impact of bone-immune system interactions on osteoporosis progression. The pathogenesis of osteoporosis is significantly influenced by T cells, particularly cytotoxic and helper T cells, which modulate osteoclast differentiation and activity. A crucial aspect of understanding osteoporosis is how T lymphocytes interact with osteoclasts. However, the precise mechanisms underlying T cell-osteoclast crosstalk remain poorly understood. This review systematically examines T cell and osteoclast involvement in osteoimmunology, with a particular focus on their involvement in osteoporosis. It seeks to elucidate the immune mechanisms driving the progression of osteoporosis and identify key molecules involved in T cell-osteoclast interactions. This aims to discover novel molecular targets and intervention strategies to improve early diagnosis and management of osteoporosis. Furthermore, this article will explore the potential of intervening in T cell-osteoclast interactions using conventional therapies, traditional Chinese medicine, immunomodulatory agents, and nanomaterial-based treatments, providing new perspectives for future osteoporosis management.
Collapse
Affiliation(s)
- Zeyao Lu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peilun Xiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shijia Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chongjun Huang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weishang Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanheng Mao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Ye Tian
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
31
|
Shi D, Li Y, Tian M, Xue M, Wang J, An H. Nanomaterials-Based Drug Delivery Systems for Therapeutic Applications in Osteoporosis. Adv Biol (Weinh) 2025:e2400721. [PMID: 40195930 DOI: 10.1002/adbi.202400721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/04/2025] [Indexed: 04/09/2025]
Abstract
The etiology of osteoporosis is rooted in the disruption of the intricate equilibrium between bone formation and bone resorption processes. Nevertheless, the conventional anti-osteoporotic medications and hormonal therapeutic regimens currently employed in clinical practice are associated with a multitude of adverse effects, thereby constraining their overall therapeutic efficacy and potential. Recently, nanomaterials have emerged as a promising alternative due to their minimal side effects, efficient drug delivery, and ability to enhance bone formation, aiding in restoring bone balance. This review delves into the fundamental principles of bone remodeling and the bone microenvironment, as well as current clinical treatment approaches for osteoporosis. It subsequently explores the research status of nanomaterial-based drug delivery systems for osteoporosis treatment, encompassing inorganic nanomaterials, organic nanomaterials, cell-mimicking carriers and exosomes mimics and emerging therapies targeting the osteoporosis microenvironment. Finally, the review discusses the potential of nanomedicine in treating osteoporosis and outlines the future trajectory of this burgeoning field. The aim is to provide a comprehensive reference for the application of nanomaterial-based drug delivery strategies in osteoporosis therapy, thereby fostering further advancements and innovations in this critical area of medical research.
Collapse
Affiliation(s)
- Donghong Shi
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, School of Electrical Engineering, Hebei University of Technology, Tianjin, 300130, P. R. China
| | - Yuling Li
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Meng Tian
- Hebei Tourism College, Hebei, Chengde, 067000, P. R. China
| | - Mengge Xue
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Jinping Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Hailong An
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| |
Collapse
|
32
|
Pi Z, Wu Y, Wang X, Li P, Wang R. Exosomal Manf originated from endothelium regulated osteoclast differentiation by down-regulating NF-κB signaling pathway. J Orthop Surg Res 2025; 20:349. [PMID: 40197525 PMCID: PMC11978012 DOI: 10.1186/s13018-025-05671-w] [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: 09/03/2024] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Endothelium-derived exosomes has been reported to enhanced osteogenesis. However, the role of endothelial exosomes on osteoclastgenesis is still unknown. METHODS Human umbilical vein endothelial cells (HUVECs) were used to isolate exosomes. PBS or HUVEC-Exos were used to treat RAW 264.7 cells. Then, the preconditioned RAW 264.7 cells were subjected to TRAP staining and RT-qPCR assays. In vivo, we constracted osteoporosis mice model. PBS or HUVEC-Exos were injected through tail vein after ovariectomy surgery. Bone mass was assessed by micro-CT and TRAP staining. Furthermore, we conducted RNA sequencing and found the genes that were differentially expressed. RESULTS Osteoclast differentiation was inhibited by endothelium-derived exosomes in this study. Moreover, HUVEC-Exos demonstrated a specific action on bones to promote in vivo bone resorption. Furthermore, exosomal Manf promoted bone resorption via down-regulating NF-κB signaling, and HUVEC-Exos Manf inhibited osteoclast differentiation in vivo. CONCLUSION HUVEC-exosomal Manf suppressed osteoclastogenesis via down-regulating NF-κB signaling.
Collapse
Affiliation(s)
- Zhilong Pi
- Guangzhou University of Chinese Medicine, No. 132, Outer Ring East road, Guangzhou, 510010, Guangdong, China
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma and Tissue Repair of Tropical Area of PLA, Hospital of Orthopaedics, General Hospital of Southern Theater Command of PLA, 111 Liuhua Road, Guangzhou, Guangdong, China
| | - You Wu
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma and Tissue Repair of Tropical Area of PLA, Hospital of Orthopaedics, General Hospital of Southern Theater Command of PLA, 111 Liuhua Road, Guangzhou, Guangdong, China
| | - Xinyu Wang
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma and Tissue Repair of Tropical Area of PLA, Hospital of Orthopaedics, General Hospital of Southern Theater Command of PLA, 111 Liuhua Road, Guangzhou, Guangdong, China
| | - Pingyue Li
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma and Tissue Repair of Tropical Area of PLA, Hospital of Orthopaedics, General Hospital of Southern Theater Command of PLA, 111 Liuhua Road, Guangzhou, Guangdong, China.
| | - Renkai Wang
- Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma and Tissue Repair of Tropical Area of PLA, Hospital of Orthopaedics, General Hospital of Southern Theater Command of PLA, 111 Liuhua Road, Guangzhou, Guangdong, China.
| |
Collapse
|
33
|
Zhang X, Yuan X, Li X, Yu H, Wang T, Zhang C, Wu J, You X. Sodium Danshensu alleviates bone cancer pain by inhibiting the osteoclast differentiation and CGRP expression. Eur J Pharmacol 2025; 992:177296. [PMID: 39900329 DOI: 10.1016/j.ejphar.2025.177296] [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: 09/12/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/05/2025]
Abstract
CONTEXT The morbidity of bone cancer pain (BCP) is on the rise, yet current treatments have limited analgesic efficacy. Sodium Danshensu (SDSS), or sodium 3-(3,4-dihydroxyphenyl)-DL-lactate, exhibits anti-inflammatory, anti-osteoporotic properties. Current research shows that bone cancer pain is closely related to the development of osteoclasts. OBJECTIVE To investigate the analgesic effects of SDSS on BCP in mice and explore the underlying mechanisms. MATERIALS & METHODS Nociceptive behaviors in BCP mice were evaluated by paw withdrawal threshold (PWT) and limb using score (LUS). Network pharmacology, Gene Ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation, and molecular docking identified potential targets. Histological analyses, Western blot, RT-qPCR, ELISA, and immunofluorescence staining were performed on mice femurs. RESULTS SDSS significantly increased PWT and LUS in BCP mice. Forty-three common targets were identified, with the estrogen signaling pathway showing the highest enrichment. Molecular docking analysis suggested a potential binding affinity between SDSS and ESRα. SDSS administration up-regulated ESRα expression and down-regulated RANKL, RANK, NFATc1, c-fos, TRAP, and Cathepsin K (CTSK). In addition, SDSS suppressed the abnormal increase of calcitonin gene-related peptide-positive (CGRP+) neural budding and expression in nerve endings, effects which were reversed by ESRa inhibitor ICI-182780. CONCLUSIONS SDSS relieves bone cancer pain by inhibiting osteoclast activity, providing a potential new drug option for cancer pain patients.
Collapse
Affiliation(s)
- Xiaoxuan Zhang
- School of Medicine, Shanghai University, Shanghai, 200444, China; Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200030, China
| | - Xinru Yuan
- School of Medicine, Shanghai University, Shanghai, 200444, China; Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200030, China
| | - Xin Li
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200030, China; School of Medical Instrument and Food Engineering USST, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Haonan Yu
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Tingfang Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Chuan Zhang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jingxiang Wu
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200030, China.
| | - Xingji You
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
34
|
Luo J, Chen K, Nong X. Potential regulation of artesunate on bone metabolism through suppressing inflammatory infiltration in type 2 diabetes mellitus. Immunopharmacol Immunotoxicol 2025; 47:147-158. [PMID: 39762719 DOI: 10.1080/08923973.2024.2444953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 12/15/2024] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Osteoimmunology is an emerging field that explores the interplay between bone and the immune system. The immune system plays a critical role in the pathogenesis of diabetes and significantly affects bone homeostasis. Artesunate, a first-line treatment for malaria, is known for its low toxicity and multifunctional properties. Increasing evidence suggests that artesunate possesses anti-inflammatory, immunoregulatory, and osteogenic effects. This review aims to explore the relationship between immune regulation and bone metabolism in type 2 diabetes (T2DM) and to investigate the potential therapeutic application of artesunate. METHODS This review systematically examines literature from PubMed/Medline, Elsevier, Web of Science, Embase, the International Diabetes Federation, and other relevant databases. RESULTS This review synthesizes evidence from multiple sources to delineate the relationship between T lymphocytes and T2DM, the regulation of T lymphocyte subsets in bone metabolism, and the effects of artesunate on both T lymphocytes and bone metabolism. Recent studies suggest a bidirectional regulatory relationship between T2DM and T lymphocytes (CD4+ T and CD8+ T) during the onset and progression of the disease, with inflammatory and anti-inflammatory cytokines serving as key mediators. T lymphocyte subsets and their cytokines play a pivotal role in regulating osteogenesis and osteoclastogenesis in pathological conditions. Furthermore, artesunate has shown promise in modulating inflammatory infiltration and bone metabolism. CONCLUSION The accumulated evidence indicates that artesunate exerts regulatory effects on bone metabolism in T2DM by influencing T lymphocyte differentiation.
Collapse
Affiliation(s)
- Jinghong Luo
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Kun Chen
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolin Nong
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
35
|
Shi D, Fernando IPS, Vahedifar A, Ghosh S, Wu J. Canola Protein Hydrolysates Show Osteogenic Activity in MC3T3-E1 Cells. J Food Sci 2025; 90:e70210. [PMID: 40260770 PMCID: PMC12012869 DOI: 10.1111/1750-3841.70210] [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: 12/17/2024] [Revised: 03/18/2025] [Accepted: 03/31/2025] [Indexed: 04/24/2025]
Abstract
Osteoporosis, the most prevalent metabolic bone disorder, is a major public health issue. Previous studies indicated the potential of food components in mitigating the risks of osteoporosis. The study aimed to evaluate the potential of canola protein hydrolysates (CPH) on osteoclastogenesis using a pre-osteoblast cell MC3T3-E1. Twenty-two CPHs were prepared by 12 different proteases, either individually or in combination. Three CPHs, prepared by trypsin (CPH-T), Protex 6L (CPH-P), and the combination of Protex 6L and thermoase (CPH-PT) showed promising activity in promoting in vitro bone formation. CPH-T and CPH-PT improved cell proliferation at a concentration of 10 ug/ml, while all three hydrolysates exhibited cytotoxicity at 1000 ug/ml. All three hydrolysates promoted the level of runt-related transcription factor 2 (RUNX2) and type I collagen, and mineralization in osteoblast cells, in a dose-dependent manner. Additionally, these three hydrolysates elevated the osteoprotegerin (OPG) level and reduced the level of receptor activator of nuclear factor kappa-B ligand (RANKL). This study indicated the activity of CPHs in the promotion of bone formation and prevention of osteoclastogenesis, suggesting the potential of CPHs as a promising functional food ingredient against osteoporosis.
Collapse
Affiliation(s)
- Da Shi
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaAlbertaCanada
| | - Ilekuttige Priyan Shanura Fernando
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaAlbertaCanada
| | - Amir Vahedifar
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaAlbertaCanada
| | - Supratim Ghosh
- Department of Food and Bioproduct Sciences, College of Agriculture and BioresourcesUniversity of SaskatchewanSaskatchewanCanada
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaAlbertaCanada
| |
Collapse
|
36
|
Ricucci D, Milovidova I, Williams RK, Tay F. Predentine is not a reliable barrier against internal root resorption in the presence of inflammation. J Dent 2025; 155:105626. [PMID: 39952548 DOI: 10.1016/j.jdent.2025.105626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025] Open
Abstract
INTRODUCTION Predentine has traditionally been perceived as a protective barrier against internal resorption in dental tissues. However, recent observations suggest that this barrier may be compromised under pathological conditions, particularly in the presence of inflammation. The present study explored the histological changes in teeth affected by internal resorption, and examined the role of predentine in resisting resorptive activity. METHODS The study utilised 44 extracted human teeth with varying degrees of caries involvement and pulpal inflammation. Longitudinal serial sections were prepared for histological examination. Haematoxylin and eosin staining was used to observe tissue morphology, while a modified Brown and Brenn staining technique was employed to identify bacteria and their infiltration status. The focus was on identifying resorptive lacunae, odontoclast activity, and the integrity of the predentine layer. RESULTS Histological analysis revealed resorptive activity in 23 out of 44 teeth. Odontoclasts were frequently observed in close contact with predentine, particularly in areas of inflammation. Despite the presence of predentine, resorption extended into the underlying mineralised dentine, challenging the notion of predentine as an impermeable barrier. Bacterial colonisation was evident in necrotic areas, correlating with regions of active resorption. CONCLUSION The findings suggest that predentine may not be as effective a barrier against internal resorption as previously thought, particularly in inflamed conditions. Future research should aim to elucidate the mechanisms underlying odontoclast activity and develop strategies to prevent or mitigate internal resorption in clinical practice. CLINICAL SIGNIFICANCE This study highlights the need for early intervention in cases of dental inflammation to prevent internal resorption, as predentine may not provide sufficient protection in compromised conditions.
Collapse
Affiliation(s)
| | | | - Reese K Williams
- Department of Endodontics, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Franklin Tay
- Department of Endodontics, Dental College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
37
|
Seo J, Ko R, Kim M, Seo J, Lee H, Kim D, Jeong W, Kim HS, Lee SY. Pim1 promotes the maintenance of bone homeostasis by regulating osteoclast function. Exp Mol Med 2025; 57:733-744. [PMID: 40164682 PMCID: PMC12046003 DOI: 10.1038/s12276-025-01421-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/24/2024] [Accepted: 01/05/2025] [Indexed: 04/02/2025] Open
Abstract
The Pim1 (proviral integration site for Moloney leukemia virus 1) protein is a serine/threonine kinase that is essential for cell proliferation, apoptosis and innate immune responses. Here we show that Pim1 promotes osteoclast resorptive function without affecting osteoclast numbers. Specifically, we found that mice lacking Pim1 (Pim1-/-) developed increased trabecular bone mass and indices such as trabecular bone-mass density. This was due to the direct phosphorylation of TRAF6 by Pim1 in mature osteoclasts, which activated the Akt-GSK3β signaling pathway. This, in turn, promoted the acetylation and consequent stabilization of microtubules, which permitted the formation of the osteoclast sealing zone. In vivo experiments then showed that, when mice with lipopolysaccharide-induced bone loss or tumor-induced osteolysis were treated with SGI-1776, a Pim1 inhibitor that is more selective for Pim1, the bone loss was significantly ameliorated. Thus, Pim1 plays an important role in osteoclast function and may be a therapeutic target for bone-related diseases.
Collapse
Affiliation(s)
- Jeongin Seo
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea
| | - Ryeojin Ko
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
| | - Minhee Kim
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea
| | - Jeongmin Seo
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Doyong Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Woojin Jeong
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Soo Young Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea.
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea.
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
38
|
Tarar A, Sanaei R, Ayodele BA, DiGiacomo K, Leury BJ, Mackie EJ, Woodward AP, Pagel CN. Changes in concentrations of cytokines and markers of bone turnover in dairy cows during different stages of a production cycle. J Dairy Sci 2025; 108:4448-4461. [PMID: 40043754 DOI: 10.3168/jds.2024-25564] [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: 08/07/2024] [Accepted: 01/23/2025] [Indexed: 04/20/2025]
Abstract
Changes in bone metabolism occur to meet the varying demands for calcium during pregnancy and lactation. These changes are regulated locally and systemically by a variety of factors, including hormones, cytokines, chemokines, and growth factors. The goal of the present experiment was to document the dynamics of bone turnover and metabolic state during a complete lactation cycle in dairy cows and to monitor the plasma profile of 10 cytokines previously known to play roles in bone turnover during lactation. Blood samples were collected sequentially from Holstein-Friesian cows during late pregnancy (1-9 wk prepartum; n = 30), early lactation (4-8 wk postpartum [pp]; n = 30), mid lactation (18-22 wk pp; n = 26), and late lactation (35-39 wk pp; n = 21). Due to reproductive failures, 8 of the cows went through extended lactation and samples for these cows were collected at a fifth time point, extended lactation (51-71 wk pp; n = 8). Twelve nonpregnant, nonlactating cows were also sampled. These cows included 4 heifers and 8 nonpregnant (empty) cows. Samples were assayed for calcium, phosphate, the bone resorption marker C-terminal cross linking telopeptide of type I collagen (CTX-I), the bone formation marker, osteocalcin, and 3 metabolic markers, nonesterified fatty acids (NEFA), BHB, and glucose. The plasma levels of IL-1β, IL-4, IL-6, IL-8, IL-10, IFN-γ, IL-17A, TNF-α, MIP-1α, and VEGF-A were also measured using a multiplex cytokine assay. Plasma calcium and phosphate concentrations were found to be within the normal range across all lactation stages. Plasma CTX-I and osteocalcin concentrations corresponded to the stage of production, showing higher levels of CTX-I in late pregnancy and early lactation, and higher levels of osteocalcin in mid lactation, than at other stages of the production cycle. Analysis of metabolic markers, NEFA, and BHB, indicated that fat metabolism was increased to meet high energy demands during specific reproductive stages. The results showed that although glucose concentrations and IL-10, TNF-α, and MIP-1α were not detectably different at any of the stages of production, IL-4, IL-6, IL-8, IFN-γ, and VEGF-A were affected by stage of production. Most of the changes observed in cytokine levels occurred during late pregnancy and late lactation. Plasma IL-6 levels were high during late pregnancy, whereas plasma levels of IFN-γ and IL-4 were elevated during late lactation. In conclusion, these data demonstrate that plasma concentrations of a biomarker bone resorption, but not bone formation, and the cytokines IL-4, IL-6, and IFN-γ are related to the stage of production in dairy cattle. Furthermore, the timing of changes in plasma concentrations of cytokines differ, with plasma IL-6 concentration elevated in parallel with CTX-I concentrations and plasma IL-4 and IFN-γ concentrations elevated later in lactation.
Collapse
Affiliation(s)
- Aisha Tarar
- Veterinary Bioscience, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Reza Sanaei
- Veterinary Bioscience, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Babatunde A Ayodele
- Veterinary Bioscience, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Kristy DiGiacomo
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brian J Leury
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Eleanor J Mackie
- Veterinary Bioscience, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andrew P Woodward
- Veterinary Bioscience, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Charles N Pagel
- Veterinary Bioscience, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
39
|
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
|
40
|
Lungu O, Toscani D, Giuliani N. Mechanistic insights into bone destruction in multiple myeloma: Cellular and molecular perspectives. J Bone Oncol 2025; 51:100668. [PMID: 40124903 PMCID: PMC11928850 DOI: 10.1016/j.jbo.2025.100668] [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: 01/28/2025] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy that leads to significant bone destruction, resulting in debilitating pain and skeletal-related events. The pathophysiology of osteolytic bone destruction in MM involves complex interactions between malignant plasma cells (PCs) and the bone marrow (BM) microenvironment. This review aims to provide a comprehensive synthesis of the cellular and molecular pathways underlying MM-associated bone disease. We discuss the role of osteoclast (OC), osteoblast (OB), osteocytes, along with the complex interactions between immune cells and the BM microenvironment in shaping disease progression. Additionally, we explore the molecular signaling pathways involved in bone disease as well as the influence of inflammatory cytokines, and the role of the metabolic alterations that characterize the MM BM. We also explore novel therapeutic strategies targeting these pathways to improve clinical outcomes. Understanding these mechanisms is crucial for the development of more effective treatments to prevent bone damage in MM patients.
Collapse
Affiliation(s)
- Oxana Lungu
- Laboratory of Hematology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Denise Toscani
- Laboratory of Hematology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Nicola Giuliani
- Laboratory of Hematology, Department of Medicine and Surgery, University of Parma, Parma, Italy
- Hematology and BMT Unit, “Azienda Ospedaliero-Universitaria di Parma”, Parma, Italy
| |
Collapse
|
41
|
Xi H, Jiang X, Xiong S, Zhang Y, Zhou J, Liu M, Zhou Z, Zhang C, Liu S, Long Z, Zhou J, Qian G, Xiong L. 3D-printed gallium-infused scaffolds for osteolysis intervention and bone regeneration. Mater Today Bio 2025; 31:101524. [PMID: 39980629 PMCID: PMC11840525 DOI: 10.1016/j.mtbio.2025.101524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/22/2025] Open
Abstract
Exacerbation of osteolysis in osteoporotic bone defects presents a significant challenge for implant-based treatments. This underscores the urgent need to develop implants that actively mitigate osteolysis while simultaneously promoting bone regeneration. In this study, the osteogenic potential of mesoporous bioactive glass (MBG) and β-tricalcium phosphate (β-TCP) was combined with the anti-bone resorption property of Ga doping. Ga-MBG was synthesized using a self-transformation method and subsequently incorporated into β-TCP at concentrations of 5 wt%, 10 wt% and 15 wt%. Scaffolds were prepared using extrusion-based 3D printing. The cytocompatibility of the composite scaffolds and their regulatory effects on the differentiation of osteoblasts and osteoclasts were systematically examined. In addition, the molecular mechanisms underlying bone regeneration and osteolysis regulation in osteoblasts were explored. Subsequently, cranial defects were repaired in a rat model of osteoporosis to assess the therapeutic efficacy and biological safety of the optimal concentration of the Ga-MBG/TCP composite scaffold. These findings indicated that the 10 wt% Ga-MBG/TCP composite scaffold exhibited excellent biocompatibility, enhanced new bone formation, and effectively mitigated osteolysis. These results provide a foundation for further investigation into the optimal concentration of Ga-MBG implants and highlight their potential application in future therapies for osteoporotic bone defects.
Collapse
Affiliation(s)
- Hanrui Xi
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, NO. 1 Minde Road, Nanchang, Jiangxi, 330006, China
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
| | - Xihao Jiang
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, No. 1180 Shuanggang East Avenue, Nanchang, Jiangxi, 330013, China
| | - Shilang Xiong
- Department of Orthopedics, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Yinuo Zhang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jingyu Zhou
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, NO. 1 Minde Road, Nanchang, Jiangxi, 330006, China
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
| | - Min Liu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, NO. 1 Minde Road, Nanchang, Jiangxi, 330006, China
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
| | - Zhigang Zhou
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, NO. 1 Minde Road, Nanchang, Jiangxi, 330006, China
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
| | - Chengyu Zhang
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, NO. 1 Minde Road, Nanchang, Jiangxi, 330006, China
| | - Shiwei Liu
- Department of Joint Surgery, Ganzhou People's Hospital, No. 16, Mei Guan Road, Zhang Gong District, Ganzhou, Jiangxi, 341000, China
| | - Zhisheng Long
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
- Department of Orthopedic, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, No. 16, Mei Guan Road, Zhang Gong District, Ganzhou, Jiangxi, 341000, China
| | - Guowen Qian
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, No. 1180 Shuanggang East Avenue, Nanchang, Jiangxi, 330013, China
| | - Long Xiong
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, NO. 1 Minde Road, Nanchang, Jiangxi, 330006, China
- Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Jiangxi, 330006, China
- Institute of Minimally Invasive Orthopedics, Nanchang University, Jiangxi, 330006, China
| |
Collapse
|
42
|
Luo F, Chen T, Chen S, Bai D, Li X. Regulation of osteoclast-mediated bone resorption by lipids. Bone 2025; 193:117423. [PMID: 39933643 DOI: 10.1016/j.bone.2025.117423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
Hyperactivation of osteoclasts has been identified as a significant etiological factor in several bone resorption-related disorders, including osteoporosis, periodontitis, arthritis, and bone metastasis of tumors. It has been demonstrated that the severity of these diseases is influenced by lipids that regulate osteoclast differentiation and activity through specific signaling pathways and cytokine levels. The regulatory mechanisms of different types of lipids on osteoclastogenesis vary across diverse disease contexts in bone resorption regulated by osteoclasts. This review presents an overview of the mechanisms underlying osteoclast formation and summarizes the pathways through which various lipids regulate osteoclastogenesis in different pathological contexts. We also discuss effective therapeutic strategies for osteolytic diseases based on modulation of lipid metabolism.
Collapse
Affiliation(s)
- Fang Luo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Tianyi Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Song Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinyi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
43
|
Shin SY, Kang IS, Kim C. ERK inhibits osteoclast differentiation in RAW 264.7 cells through the osteoprotegerin-mediated autophagy. Bone 2025; 193:117424. [PMID: 39947572 DOI: 10.1016/j.bone.2025.117424] [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: 11/05/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
Osteoclasts (OCs) are bone-resorbing cells derived from the monocyte/macrophage lineage. The extracellular signal-regulated kinase (ERK) pathway controls cellular responses such as proliferation, differentiation, and survival, including those of OCs. In the present study, ERK inhibitors reduced the proliferation of bone marrow-derived macrophages (BMMs) and RAW 264.7 cells. However, ERK inhibitors decreased OC differentiation in BMMs but increased it in RAW 264.7 cells. ERK downregulation using small interfering RNA transfection also increased the OC differentiation and the expression of receptor activator of nuclear factor-κB, OC-specific markers, and OC-associated transcription factors in RAW 264.7 cells. These findings suggest ERK regulates OC differentiation in RAW 264.7 cells differently than in BMMs. Thus, we further investigated the mechanism by which ERK negatively regulates OC differentiation in RAW 264.7 cells. ERK inhibition decreased the expression of osteoprotegerin (OPG), a negative regulator of OC differentiation. OPG knockdown increased OC formation. ERK inhibitors activated the Akt/mammalian target of the rapamycin (mTOR) signaling pathway while inhibiting unc-51-like autophagy activating kinase 1 (ULK1). This resulted in decreased levels of microtubule-associated protein 1A/1B-light chain 3-II (LC3-II) and increased levels of p62, thereby reducing autophagy. In addition, OPG knockdown reduced autophagy by activating Akt/mTOR and inhibiting ULK1, resulting in decreased LC3-II and accumulated p62. Therefore, ERK inhibition promoted OC differentiation by downregulating OPG-mediated inhibition of osteoclastogenesis and autophagy in RAW 264.7 cells. These findings highlight ERK's complex role in OC differentiation and suggest that understanding ERK's dual impact on OC differentiation can provide insights into novel treatment strategies for bone-related disorders.
Collapse
Affiliation(s)
- Soo-Young Shin
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea; BK21, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - In-Soon Kang
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Chaekyun Kim
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea; BK21, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
44
|
Chen Y, Zou J, Niimi M, Qiu X, Zhang S, Yang H, Zhu M, Fan J. Matrix Metalloproteinase-9 Enhances Osteoclastogenesis: Insights from Transgenic Rabbit Bone Marrow Models and In Vitro Studies. Int J Mol Sci 2025; 26:3194. [PMID: 40244002 PMCID: PMC11989254 DOI: 10.3390/ijms26073194] [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: 02/13/2025] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Osteoclastogenesis is tightly regulated by receptor activator of nuclear factor kappa-B ligand (RANKL) signaling, yet the role of matrix metalloproteinase-9 (MMP-9) in this process remains controversial. We established a high-yield osteoclastogenesis system using cryopreserved rabbit bone marrow cells (1 × 109 cells/femur) treated with Macrophage colony-stimulating factor (M-CSF) and RANKL. Bone marrow cells from MMP-9 transgenic rabbits (macrophage-specific overexpression) and MMP-9-transfected RAW264.7 macrophages were compared to wild-type controls. MMP-9 overexpression increased osteoclastogenesis 5.5-fold (20 ng/mL RANKL, * p < 0.01) while suppressing inflammatory cytokines (IL-1β, TNF-α). RAW264.7 macrophages stably transfected with human MMP-9 similarly exhibited reduced inflammatory cytokine levels and enhanced osteoclastogenesis. MMP-9 acts as a dual regulator of osteoclastogenesis and inflammation, suggesting therapeutic potential for osteoporosis management.
Collapse
Affiliation(s)
- Yajie Chen
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China; (Y.C.); (J.Z.); (X.Q.); (S.Z.); (H.Y.)
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan;
| | - Jialun Zou
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China; (Y.C.); (J.Z.); (X.Q.); (S.Z.); (H.Y.)
| | - Manabu Niimi
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan;
| | - Xuan Qiu
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China; (Y.C.); (J.Z.); (X.Q.); (S.Z.); (H.Y.)
| | - Shuang Zhang
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China; (Y.C.); (J.Z.); (X.Q.); (S.Z.); (H.Y.)
| | - Han Yang
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China; (Y.C.); (J.Z.); (X.Q.); (S.Z.); (H.Y.)
| | - Maobi Zhu
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China; (Y.C.); (J.Z.); (X.Q.); (S.Z.); (H.Y.)
| | - Jianglin Fan
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China; (Y.C.); (J.Z.); (X.Q.); (S.Z.); (H.Y.)
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan;
| |
Collapse
|
45
|
Liu L, Zhang J, Cui R, Wang N, Zhang Y, Liu L, Zhang X, Liu Q. SIRT1 and exercise-induced bone metabolism: a regulatory nexus. Front Cell Dev Biol 2025; 13:1522821. [PMID: 40206398 PMCID: PMC11979185 DOI: 10.3389/fcell.2025.1522821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025] Open
Abstract
Regular exercise positively influences bone health, enhances bone density and strength, and reduces the risk of osteoporosis. Silent information regulator of transcription 1 (SIRT1) is a deacetylase that plays a pivotal role in the regulation of various biological processes. In this review, we explore the role of SIRT1 in modulating bone metabolism in response to exercise. SIRT1 regulates crucial cellular processes, including inflammation, aging, autophagy, and oxidative stress, in bone cells such as bone marrow mesenchymal stem cells, osteoblasts, and osteoclasts, in response to exercise-induced stimuli. Notably, exercise influences bone metabolism by modulating muscle metabolism and neurotransmitters, with SIRT1 acting as a key mediator. A comprehensive understanding of SIRT1's regulatory mechanisms will facilitate a deeper exploration of the principles underlying exercise-induced improvements in bone metabolism, ultimately providing novel insights into the treatment of bone metabolic disorders.
Collapse
Affiliation(s)
- Lijie Liu
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Jiale Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Runhong Cui
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Na Wang
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Yun Zhang
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Lifei Liu
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Xinan Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Qingfeng Liu
- Department of General Surgery, Jinqiu Hospital of Liaoning Province, Shenyang, China
| |
Collapse
|
46
|
Dey G, Yakobovich E, Loboda J, Sinai-Turyansky R, Abramovitch-Dahan C, Merquiol E, Sridharan N, Itzhak G, Turk B, Wald O, Turk D, Yona S, Levaot N, Blum G. Development and Application of Small Molecule-Peptide Conjugates as Cathepsin K-Specific Covalent Irreversible Inhibitors in Human Osteoclast and Lung Cancer. JACS AU 2025; 5:1104-1120. [PMID: 40151260 PMCID: PMC11938014 DOI: 10.1021/jacsau.4c00840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 03/29/2025]
Abstract
Cathepsin K (CTSK), a proteolytic enzyme that degrades the extracellular matrix, is recognized as a significant therapeutic target for osteoporosis, osteoarthritis, and rheumatoid arthritis. Due to adverse effects, no clinically approved drugs exist for CTSK. In order to develop safer therapeutics, highly selective CTSK inhibitors are required to elucidate the origins of side effects. Here, we developed various hybrid inhibitors by combining peptide sequences with small organic molecules. An acyloxymethyl ketone electrophile was incorporated as a bioisostere of the glycine-glycine cleavage site and inverse peptide sequences to enhance prime site interactions, as seen in the crystal structure. Additionally, a diphenyl group was incorporated to improve nonprime site interactions, culminating in highly selective and potent irreversible CTSK inhibitors with negligible off-target binding by closely related cathepsins. These novel inhibitors were also designed to attach to targeting moieties, further reducing off-target effects in vivo. Our findings demonstrate that these highly selective inhibitors are nontoxic, effectively inhibit bone resorption by human osteoclasts, block CTSK activity in cells and their nuclei, and inhibit activity in human lung cancer tissue. This study highlights significant advancements in designing CTSK inhibitors with potential clinical applications for lung cancer and osteoclast-related conditions.
Collapse
Affiliation(s)
- Gourab Dey
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, Jerusalem 9112001, Israel
| | - Evalyn Yakobovich
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, Jerusalem 9112001, Israel
| | - Jure Loboda
- Department
of Biochemistry and Molecular Biology, J.
Stefan Institute, Jamova 39, SI-1000 Ljubljana, Sloveni
| | - Reut Sinai-Turyansky
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, Jerusalem 9112001, Israel
| | - Chen Abramovitch-Dahan
- Department
of Physiology and Cell Biology Faculty of Health Sciences, Ben-Gurion University of the Negev, Shderot Ben Gurion 1, Beer-Sheva 844394, Israel
| | - Emmanuelle Merquiol
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, Jerusalem 9112001, Israel
| | - Nikhila Sridharan
- The
Institute of Biomedical and Oral Research, The Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Gal Itzhak
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, Jerusalem 9112001, Israel
| | - Boris Turk
- Department
of Biochemistry and Molecular Biology, J.
Stefan Institute, Jamova 39, SI-1000 Ljubljana, Sloveni
| | - Ori Wald
- Department
of Cardiothoracic Surgery, Hadassah Hebrew University Medical Center,
The Faculty of Medicine, The Hebrew University
of Jerusalem, Jerusalem 9112001, Israel
| | - Dusan Turk
- Department
of Biochemistry and Molecular Biology, J.
Stefan Institute, Jamova 39, SI-1000 Ljubljana, Sloveni
| | - Simon Yona
- The
Institute of Biomedical and Oral Research, The Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Noam Levaot
- Department
of Physiology and Cell Biology Faculty of Health Sciences, Ben-Gurion University of the Negev, Shderot Ben Gurion 1, Beer-Sheva 844394, Israel
| | - Galia Blum
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, Jerusalem 9112001, Israel
| |
Collapse
|
47
|
Ramani H, Cleret-Buhot A, Sylla M, Bunet R, Bertrand F, Peet MM, Chartrand-Lefebvre C, Trottier B, Thomas R, Routy JP, Fortin C, Martel-Laferrière V, Sadouni M, Cloutier G, Allard L, Kizer JR, Chomont N, Ancuta P, Hanna DB, Kaplan RC, Jenabian MA, Landay AL, Durand M, El-Far M, Tremblay CL. Opposite Roles of IL-32α Versus IL-32β/γ Isoforms in Promoting Monocyte-Derived Osteoblast/Osteoclast Differentiation and Vascular Calcification in People with HIV. Cells 2025; 14:481. [PMID: 40214435 PMCID: PMC11987946 DOI: 10.3390/cells14070481] [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: 02/01/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
People with HIV (PWH) have an increased risk of developing cardiovascular disease (CVD). Our recent data demonstrated that the multi-isoform proinflammatory cytokine IL-32 is upregulated in PWH and is associated with arterial stiffness and subclinical atherosclerosis. However, the mechanisms by which IL-32 contributes to the pathogenesis of these diseases remain unclear. Here, we show that while the less expressed IL-32α isoform induces the differentiation of human classical monocytes into the calcium-resorbing osteoclast cells, the dominantly expressed isoforms IL-32β and IL-32γ suppress this function through the inhibition of TGF-β and induce the differentiation of monocytes into the calcium-depositing osteocalcin+ osteoblasts. These results aligned with the increase in plasma levels of osteoprotegerin, a biomarker of vascular calcification, and its association with the presence of coronary artery subclinical atherosclerosis and calcium score in PWH. These findings support a novel role for the proinflammatory cytokine IL-32 in the pathophysiology of CVD by increasing vascular calcification in PWH.
Collapse
Affiliation(s)
- Hardik Ramani
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada; (H.R.); (R.B.); (F.B.); (C.F.); (V.M.-L.); (N.C.); (P.A.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - Aurélie Cleret-Buhot
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
- Cellular Imaging Core Facility, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Mohamed Sylla
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - Rémi Bunet
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada; (H.R.); (R.B.); (F.B.); (C.F.); (V.M.-L.); (N.C.); (P.A.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - Florent Bertrand
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada; (H.R.); (R.B.); (F.B.); (C.F.); (V.M.-L.); (N.C.); (P.A.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - Marc-Messier Peet
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - Carl Chartrand-Lefebvre
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
- Département de Radiologie, Radio-Oncologie et Médecine Nucléaire, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Benoit Trottier
- Centre de Médecine Urbaine du Quartier Latin, Montréal, QC H2L 0B1, Canada;
| | - Réjean Thomas
- Clinique Médicale l’Actuel, Montréal, QC H2L 4P9, Canada;
| | - Jean-Pierre Routy
- Research Institute of McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
| | - Claude Fortin
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada; (H.R.); (R.B.); (F.B.); (C.F.); (V.M.-L.); (N.C.); (P.A.)
| | - Valérie Martel-Laferrière
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada; (H.R.); (R.B.); (F.B.); (C.F.); (V.M.-L.); (N.C.); (P.A.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - Manel Sadouni
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
- Cellular Imaging Core Facility, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Guy Cloutier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
- Département de Radiologie, Radio-Oncologie et Médecine Nucléaire, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Louise Allard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - Jorge R. Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, and Department of Medicine, Epidemiology and Biostatistics, the University of California San Francisco, San Francisco, CA 94121, USA;
| | - Nicolas Chomont
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada; (H.R.); (R.B.); (F.B.); (C.F.); (V.M.-L.); (N.C.); (P.A.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - Petronela Ancuta
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada; (H.R.); (R.B.); (F.B.); (C.F.); (V.M.-L.); (N.C.); (P.A.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - David B. Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (D.B.H.); (R.C.K.)
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (D.B.H.); (R.C.K.)
- Divsion of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada;
| | - Alan L. Landay
- Department of Internal Medicine and Microbiology and Immunology, University of Texas, Medical Branch, Austin, TX 77555, USA;
| | - Madeleine Durand
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Mohamed El-Far
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| | - Cécile L. Tremblay
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada; (H.R.); (R.B.); (F.B.); (C.F.); (V.M.-L.); (N.C.); (P.A.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (A.C.-B.); (M.S.); (M.-M.P.); (C.C.-L.); (M.S.); (G.C.); (L.A.); (M.D.)
| |
Collapse
|
48
|
Cai C, Jiang J, Li S, Gao C, Pu H, Zhao L, Xiao J. PKM2 regulates osteoclastogenesis by affecting osteoclast precursor cell fusion via downregulation of OC-STAMP and DC-STAMP. J Biol Chem 2025; 301:108439. [PMID: 40122175 DOI: 10.1016/j.jbc.2025.108439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Osteoporosis is a common bone disease that has become a serious public health problem with the aging of population. Osteoclasts are the only cells in body that can resorb bone, whose dysfunction is closely related to osteoporosis. Pyruvate kinase M2 (PKM2) is one of the essential rate-limiting enzymes in the process of glycolysis. This study aimed to elucidate the role of PKM2 in osteoclastogenesis and bone resorption. Bone marrow-derived macrophages were transfected with adenovirus to knock down the expression of PKM2 gene or treated with the PKM2 activators, DASA-58 and TEPP-46. Osteoclast formation was detected by tartrate-resistant acid phosphatase staining, osteoclast-specific gene and protein expression was detected by RT-quantitative PCR and Western blotting, and the effect of DASA-58 on osteoclast gene expression at the transcriptional level was examined by RNA sequencing. The results showed that knockdown of PKM2 by adenoviral transfection or treatment with PKM2 activators, DASA-58 and TEPP-46, inhibited osteoclast differentiation and suppressed the expression of osteoclast-associated genes in bone marrow-derived macrophages. Furthermore, PKM2 activators, DASA-58 and TEPP-46, could inhibit several signaling pathways in osteoclasts; knockdown of PKM2 or treatment with PKM2 activators, DASA-58 and TEPP-46, both affected osteoclast precursor cell fusion by inhibiting the expression of osteoclast stimulatory transmembrane protein (OC-STAMP) and dendritic cell-specific transmembrane protein (DC-STAMP). Therefore, PKM2 is closely related to osteoclast differentiation and formation, and the development of new therapeutic strategies targeting the PKM2 gene in osteoclasts may be feasible for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Cong Cai
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Jiang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenghao Gao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongxu Pu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Libo Zhao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
49
|
Liu P, Deng J, Yang Y, Bai W, Dong S, Zhang Z. Mycobacterium tuberculosis specific protein Rv1509 modulates osteoblast and osteoclast differentiation via TLR2 signaling. iScience 2025; 28:112107. [PMID: 40129707 PMCID: PMC11931388 DOI: 10.1016/j.isci.2025.112107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/09/2024] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M.tb), is one of the most ancient diseases recorded. In cases of bone TB, it significantly disrupts bone homeostasis, though the precise mechanisms are poorly understood and effective treatment targets are scarce. Our study investigated the role of Rv1509 in the pathogenesis of bone TB. We found that Rv1509 enhances the differentiation of bone marrow macrophages (BMMs) into osteoclasts by activating the TLR2 pathway, which stimulates the production of IL-6 and TNF-α. This, in turn, indirectly inhibits osteoblast differentiation and mineralization. Additionally, Rv1509 directly impairs osteoblast function and enhances the secretion of RANKL via TLR2 signaling, creating a detrimental RANKL/OPG imbalance that promotes osteoclast differentiation and bone degradation. Notably, the injection of Rv1509 into mouse skulls led to extensive bone damage, highlighting its significant role as a virulence factor in the pathogenesis of bone TB.
Collapse
Affiliation(s)
- Pan Liu
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Jiezhong Deng
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Yusheng Yang
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Wenxi Bai
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Shengtao Dong
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Zehua Zhang
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| |
Collapse
|
50
|
Zhao W, Qian J, Li J, Su T, Deng X, Fu Y, Liang X, Cui H. From death to birth: how osteocyte death promotes osteoclast formation. Front Immunol 2025; 16:1551542. [PMID: 40165960 PMCID: PMC11955613 DOI: 10.3389/fimmu.2025.1551542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Bone remodeling is a dynamic and continuous process involving three components: bone formation mediated by osteoblasts, bone resorption mediated by osteoclasts, and bone formation-resorption balancing regulated by osteocytes. Excessive osteocyte death is found in various bone diseases, such as postmenopausal osteoporosis (PMOP), and osteoclasts are found increased and activated at osteocyte death sites. Currently, apart from apoptosis and necrosis as previously established, more forms of cell death are reported, including necroptosis, ferroptosis and pyroptosis. These forms of cell death play important role in the development of inflammatory diseases and bone diseases. Increasing studies have revealed that various forms of osteocyte death promote osteoclast formation via different mechanism, including actively secreting pro-inflammatory and pro-osteoclastogenic cytokines, such as tumor necrosis factor alpha (TNF-α) and receptor activator of nuclear factor-kappa B ligand (RANKL), or passively releasing pro-inflammatory damage associated molecule patterns (DAMPs), such as high mobility group box 1 (HMGB1). This review summarizes the established and potential mechanisms by which various forms of osteocyte death regulate osteoclast formation, aiming to provide better understanding of bone disease development and therapeutic target.
Collapse
Affiliation(s)
- Weijie Zhao
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Jiale Qian
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Ji Li
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Tian Su
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, College of pharmacy, Hainan Medical University, Haikou, China
| | - Xiaozhong Deng
- Department of Pain Treatment, Nanxi Shan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Yonghua Fu
- Department of Hand and Foot Microsurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xuelong Liang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hongwang Cui
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|