1
|
Zhang Y, Cui Y, Sun C, Guo J, Li M. ED-71 ameliorates OVX-induced osteoporosis by regulating calcium homeostasis and SIRT1-mediated mitochondrial function, alleviating osteoblast senescence and suppressing osteoclastogenesis. Cell Signal 2025; 131:111713. [PMID: 40049265 DOI: 10.1016/j.cellsig.2025.111713] [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: 11/27/2024] [Revised: 02/12/2025] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Osteoporosis arising from estrogen deficiency is characterized by oxidative stress and cellular senescence accompanied by calcium loss and disrupted bone metabolism. The paracrine interaction between osteoblasts and osteoclasts, along with the ratio of receptor activator of nuclear factor-κB ligand (RANKL) to osteoprotegerin (OPG), play a pivotal role in maintaining bone homeostasis. Eldecalcitol (ED-71), a novel active form of vitamin D, can reduce the ratio of RANKL to OPG in osteoblasts. In this study, an ovariectomized (OVX) rat model was established in vivo, and a cell model was constructed in vitro using H₂O₂ to explore the specific mechanism by which ED-71 improved the release of RANKL/OPG in senescent osteoblasts. Mitochondrial dysfunction and calcium imbalance were identified as significant factors. Under oxidative stress conditions, ED-71 alleviated endoplasmic reticulum (ER) stress by decreasing the ratio of phosphorylated protein kinase R-like ER kinase (P-PERK/PERK), and augmented the expression levels of sarcoplasmic reticulum/endoplasmic reticulum calcium ATPase 2 (SCERA2) thereby promoting calcium uptake by the ER, enhancing ER calcium influx, and effectively ameliorating calcium homeostasis between the ER and mitochondria. Consequently, it mitigates mitochondrial calcium overload and associated dysfunction. In contrast, ED-71 increased the expression of silent information regulator 1 (SIRT1) and phosphorylated AMP-activated protein kinase (P-AMPK). This alleviates mitochondrial dysfunction and promotes adenosine triphosphate (ATP). The combined effects of these two factors synergistically contribute to the improvement in osteoblast senescence.
Collapse
Affiliation(s)
- Yaoguang Zhang
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China; Center of Osteoporosis and Bone Mineral Research, Shandong University, China
| | - Yajun Cui
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Center of Osteoporosis and Bone Mineral Research, Shandong University, China
| | - Changyun Sun
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China; Center of Osteoporosis and Bone Mineral Research, Shandong University, China.
| | - Jie Guo
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China.
| | - Minqi Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, China; Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China; Center of Osteoporosis and Bone Mineral Research, Shandong University, China; School of Clinical Medicine, Jining Medical University, Jining, China.; Institute of Oral Basic Research, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, China
| |
Collapse
|
2
|
Shao Z, Wang T, Yan X, Ning R, Xu X, He Q, Zhang X, Jiang M, Yang C. Identification of a RANKL/TNF-α Dual-Inhibitor as a Potential Disease-Modifying Agent for the Treatment of Knee Osteoarthritis. J Med Chem 2025. [PMID: 40358029 DOI: 10.1021/acs.jmedchem.5c00394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Osteoarthritis (OA) is a multifactorial degenerative disease involved subchondral bone remodeling, cartilage destruction and synovium inflammation. While receptor activator of nuclear factor-κB ligand (RANKL), a tumor necrosis factor (TNF) superfamily protein, is the critical regulator in bone metabolism associated with subchondral bone resorption, TNF-α is also an important inflammatory factor involved in the OA inflammation and cartilage destruction. Based on previous compound Y1599, we identified a novel tetrahydro-β-carboline derivative Y2641 with both RANKL and TNF-α inhibition in this study. Y2641 exhibited potent RANKL-induced osteoclastogenic inhibition (IC50 = 109.1 nM), and had anti-inflammatory and cartilage destruction inhibiting effects at 10 μM with low cytotoxicity. SPR assays demonstrated the binding affinity of Y2641 to RANKL (Kd = 3.984 μM) and TNF-α (Kd = 18.59 μM). In vivo assay further revealed the disease-modifying effects of Y2641 in OA rats, establishing Y2641 as a promising lead compound for the development of disease-modifying osteoarthritis drugs.
Collapse
Affiliation(s)
- Zhengguang Shao
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 200021, China
| | - Tianqi Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xueming Yan
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruonan Ning
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xing Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian He
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaofei Zhang
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Min Jiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chunhao Yang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 200021, China
| |
Collapse
|
3
|
Deng M, Ding H, Zhou Y, Qi G, Gan J. Cancer metastasis to the bone: Mechanisms and animal models (Review). Oncol Lett 2025; 29:221. [PMID: 40103600 PMCID: PMC11916653 DOI: 10.3892/ol.2025.14967] [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: 10/30/2024] [Accepted: 02/14/2025] [Indexed: 03/20/2025] Open
Abstract
The majority of cancer-related deaths result from tumor metastasis, with bone metastasis occurring in almost all types of malignant tumors. Understanding the mechanism by which tumors metastasize to bone is critical for the identification of novel therapeutic targets. A large amount of research has been carried out using animal models, and these models have been crucial in advancing the fundamental understanding of cancer. However, current models are limited; although they can mimic specific stages of the metastatic process, they are not able to replicate the entire process from tumorigenesis to bone metastasis. The present review describes the molecular changes that occur in the intraosseous microenvironment of bone metastases, including osteolytic and osteoblastic types, and summarizes advancements in animal models of bone metastasis.
Collapse
Affiliation(s)
- Meimei Deng
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi 541199, P.R. China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi 541199, P.R. China
| | - Hao Ding
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yuru Zhou
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi 541199, P.R. China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi 541199, P.R. China
| | - Guangying Qi
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi 541199, P.R. China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi 541199, P.R. China
| | - Jinfeng Gan
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi 541199, P.R. China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi 541199, P.R. China
| |
Collapse
|
4
|
Li H, Liu H, Zhou Y, Cheng L, Wang B, Ma J. The multifaceted roles of extracellular vesicles in osteonecrosis of the femoral head. J Orthop Translat 2025; 52:70-84. [PMID: 40256260 PMCID: PMC12008682 DOI: 10.1016/j.jot.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a severe disease characterized by bone tissue necrosis due to vascular impairment, often leading to joint collapse and requiring surgical intervention. Extracellular vesicles (EVs) serve as crucial mediators of intercellular communication, influencing osteogenesis, angiogenesis, and immune regulation. This review summarizes the dual role of EVs in both the pathogenesis of ONFH and post-necrosis bone repair, highlighting the impact of various EV-mediated signaling pathways on bone regeneration and the potential crosstalk among these pathways. Additionally, EVs hold promise as diagnostic biomarkers or contrast agents to complement conventional imaging techniques for ONFH detection. By elucidating the role of EVs in osteonecrosis and addressing the current challenges, we aspire to establish a foundation for the timely identification and treatment of ONFH. The translational potential of this article: This review comprehensively discusses the role of EVs in ONFH, providing innovative and promising insights for its diagnosis and treatment, which also establishes a theoretical foundation for the future clinical application of EVs in ONFH.
Collapse
Affiliation(s)
- Hongxu Li
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Haoyang Liu
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Yu Zhou
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Liming Cheng
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Bailiang Wang
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jinhui Ma
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029, China
| |
Collapse
|
5
|
Xu C, Qiu S, Yuan Z, Qiu C, Xu W, Guo J, Wen G, Liu S, Yan W, Xu H, Hou H, Yang D. Biomimetic Microstructured Scaffold with Release of Re-Modified Teriparatide for Osteoporotic Tendon-to-Bone Regeneration via Balancing Bone Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500144. [PMID: 40091692 DOI: 10.1002/advs.202500144] [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: 01/03/2025] [Revised: 02/11/2025] [Indexed: 03/19/2025]
Abstract
Osteoporotic tendon-to-bone interface healing is challenging, with a high surgical repair failure rate of up to 68%. Conventional tissue engineering approaches have primarily focused on promoting interface healing by stimulating regeneration in either the tendon or bone. However, these methods often fall short of achieving optimal therapeutic outcomes due to their neglect of balancing bone homeostasis and remodeling the microstructure at the osteoporotic tendon-to-bone interface. Herein, a series of site-specific functional modifications are carried out on teriparatide to develop recombinant human parathyroid hormone (R-PTH). A biomimetic microstructured reconstruction scaffold (BMRP) is constructed using a decalcified mussel shell scaffold, pre-gel, and R-PTH. The BMRP mimics the microstructures of the native tendon-to-bone interface and restores the original structure of the interface tissue by repairing injured cells, balancing bone homeostasis, and remodeling the microstructure of the osteoporotic tendon-to-bone interface. In an osteoporotic rotator cuff tear model, BMRP is in situ implanted at the injured site, resulting in structural reconstruction and functional recovery. The BMRP demonstrates excellent repair effects, representing a novel therapeutical alternative for treating osteoporotic tendon-to-bone injury potential for clinical application.
Collapse
Affiliation(s)
- Chengzhong Xu
- Department of Orthopaedics-Spine Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, P. R. China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Sijie Qiu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Zhigen Yuan
- Department of Orthopaedics-Spine Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, P. R. China
| | - Chongyin Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Wenyu Xu
- Department of Orthopaedics-Spine Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, P. R. China
| | - Jialiang Guo
- Department of Orthopaedics-Spine Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, P. R. China
| | - Gen Wen
- Department of Orthopaedics-Spine Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, P. R. China
| | - Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Wenjuan Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Haibing Xu
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Dehong Yang
- Department of Orthopaedics-Spine Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, P. R. China
| |
Collapse
|
6
|
Ibne Mahbub MS, Park M, Park SS, Won MJ, Lee BR, Kim HD, Lee BT. dECM and β-TCP incorporation effect on the highly porous injectable bio-glass bead for enhanced bone regeneration: In-vitro, in-vivo insights. Int J Biol Macromol 2025; 305:141040. [PMID: 39978514 DOI: 10.1016/j.ijbiomac.2025.141040] [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: 11/11/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
This study presents the development of an innovative injectable bioactive material, BG-ETa, for bone regeneration. Porcine-derived dermal extracellular matrix (dECM) was decellularized and combined with beta-tri calcium phosphate (β-TCP) and porous bio-glass (BG) beads, followed by freeze-drying to produce surface-modified BG beads. Incorporating sodium alginate (SA) enhanced injectability of the system, enabling effective delivery to defect sites. Bio-glass promotes osteogenic support and osteogenesis. dECM, rich in essential proteins and growth factors, mimics the bone microenvironment to improve cell adhesion, proliferation, and differentiation. The bioactive dECM/β-TCP coating on the bead surface offers neovascularization and early mineralization properties which ultimately facilitates new bone formation. In vitro assays demonstrated BG-ETa's biocompatibility, antimicrobial properties, and potential for osteogenic differentiation, with significant results in alkaline phosphatase (ALP) activity, alizarin red staining (ARS), immunocytochemistry (ICC), and gene expression through real-time PCR. In vivo implantation in rabbit femoral defects revealed promising degradation and significant bone regeneration after 4 and 8 weeks, as observed by histological analysis and micro-CT imaging. This injectable BG-ETa system holds promise as an effective alternative to traditional grafts, providing bioactive environment for enhanced bone regeneration with the potential to overcome limitations associated with autologous or allogeneic grafting.
Collapse
Affiliation(s)
- Md Sowaib Ibne Mahbub
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Myeongki Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Seong-Su Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Mi Jin Won
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | | | - Hai-Doo Kim
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, South Korea; Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, South Korea.
| |
Collapse
|
7
|
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
|
8
|
Mohanty S, Sahu A, Mukherjee T, Kispotta S, Mal P, Gupta M, Ghosh JK, Prabhakar PK. Molecular mechanisms and treatment strategies for estrogen deficiency-related and glucocorticoid-induced osteoporosis: a comprehensive review. Inflammopharmacology 2025:10.1007/s10787-025-01749-3. [PMID: 40293652 DOI: 10.1007/s10787-025-01749-3] [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: 12/08/2024] [Accepted: 04/04/2025] [Indexed: 04/30/2025]
Abstract
Osteoporosis, a debilitating condition characterized by reduced bone mass and increased fracture risk, is notably influenced by estrogen deficiency and glucocorticoid treatment. This comprehensive review elucidates the molecular mechanisms underpinning estrogen deficiency-related osteoporosis (EDOP) and glucocorticoid-induced osteoporosis (GIOP). The role of estrogen in bone metabolism is critically examined, highlighting its regulatory effects on bone turnover and formation through various signaling pathways. Conversely, this review explores how glucocorticoids disrupt bone homeostasis, focusing on their impact on osteoclast and osteoblast function and the subsequent alteration of bone remodeling processes. The pathogenesis of both conditions is intertwined, with estrogen receptor signaling pathways and the role of inflammatory cytokines being pivotal in driving bone loss. A detailed analysis of pathogenetic and risk factors associated with EDOP and GIOP is presented, including lifestyle and genetic factors contributing to disease progression. Modern therapeutic approaches emphasize pharmacologic, non-pharmacologic, and herbal treatments for managing EDOP and GIOP. In summary, current therapeutic strategies highlight the efficacy and the safety of various interventions. This review concludes with future directions for research, suggesting a need for novel treatment modalities and a deeper understanding of the underlying mechanisms of osteoporosis. By addressing the multifaceted nature of EDOP and GIOP, this work aims to provide insights into developing targeted therapeutic strategies and improving patient outcomes in osteoporosis management.
Collapse
Affiliation(s)
- Satyajit Mohanty
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India.
| | - Anwesha Sahu
- Division of Pharmacology, Faculty of Medical Science and Research, Sai Nath University, Ranchi, 835219, Jharkhand, India
| | - Tuhin Mukherjee
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India.
| | - Sneha Kispotta
- School of Pharmaceutical Sciences, Siksha O Anusandhan deemed to be University, Bhubaneswar, 751030, Odisha, India
| | - Payel Mal
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Muskan Gupta
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Jeet Kumar Ghosh
- Department of Pharmacy, Usha Martin University, Ranchi, 835103, Jharkhand, India
| | | |
Collapse
|
9
|
Li S, Cai X, Guo J, Li X, Li W, Liu Y, Qi M. Cell communication and relevant signaling pathways in osteogenesis-angiogenesis coupling. Bone Res 2025; 13:45. [PMID: 40195313 PMCID: PMC11977258 DOI: 10.1038/s41413-025-00417-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
Osteogenesis is the process of bone formation mediated by the osteoblasts, participating in various bone-related physiological processes including bone development, bone homeostasis and fracture healing. It exhibits temporal and spatial interconnectivity with angiogenesis, constructed by multiple forms of cell communication occurring between bone and vascular endothelial cells. Molecular regulation among different cell types is crucial for coordinating osteogenesis and angiogenesis to facilitate bone remodeling, fracture healing, and other bone-related processes. The transmission of signaling molecules and the activation of their corresponding signal pathways are indispensable for various forms of cell communication. This communication acts as a "bridge" in coupling osteogenesis to angiogenesis. This article reviews the modes and processes of cell communication in osteogenesis-angiogenesis coupling over the past decade, mainly focusing on interactions among bone-related cells and vascular endothelial cells to provide insights into the mechanism of cell communication of osteogenesis-angiogenesis coupling in different bone-related contexts. Moreover, clinical relevance and applications are also introduced in this review.
Collapse
Affiliation(s)
- Shuqing Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Xinjia Cai
- Central Laboratory, Peking University School and Hospital for Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Jiahe Guo
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Xiaolu Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Wen Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yan Liu
- Central Laboratory, Peking University School and Hospital for Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Mengchun Qi
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China.
| |
Collapse
|
10
|
Zheng H, Liu X, Liang X, Guo S, Qin B, Liu EH, Duan JA. Mechanisms and structure-activity relationships of natural polysaccharides as potential anti-osteoporosis agents: A review. Int J Biol Macromol 2025; 298:139852. [PMID: 39814301 DOI: 10.1016/j.ijbiomac.2025.139852] [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: 11/26/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
In recent years, polysaccharides derived from natural sources have garnered significant attention due to their safety and potential anti-osteoporotic effects. This review provides a comprehensive overview of the sources, distribution, structures, and mechanisms of anti-osteoporosis polysaccharides, as well as an investigation into their structure-activity relationships. Over thirty distinct, homogenous polysaccharides with anti-osteoporosis properties have been extracted from natural sources, primarily categorized as glucans, fructans, galactomannans, glucomannans, and various other heteropolysaccharides. Natural polysaccharides can effectively enhance osteoblast differentiation and mineralization while suppressing osteoclast activation, with the mechanism regulated by the BMP/SMAD/RUNX2, Wnt/Catenin, OPG/RANKL/RANK, and TLR2/NF-κB/NFATc1 signaling pathways. Furthermore, polysaccharides contribute to the prevention of osteoporosis by mitigating oxidative stress, decreasing inflammation, and modulating the gut microbiota. This review also summarizes the relationship between the monosaccharide composition, molecular weight, and glycosidic bond type of polysaccharides and their anti-osteoporotic activity. A comprehensive summary and analysis of the existing deficiencies and challenges in the research of anti-osteoporotic polysaccharides is also concluded. This review may serve as a significant reference for the discovery and utilization of naturally derived anti-osteoporotic polysaccharides in the pharmaceutical and health industries.
Collapse
Affiliation(s)
- Huili Zheng
- Nanjing University of Chinese Medicine/National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210029, China; Jiangsu Province Key Laboratory of High Technology Research, Nanjing 210029, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China
| | - Xinhui Liu
- Nanjing University of Chinese Medicine/National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210029, China; Jiangsu Province Key Laboratory of High Technology Research, Nanjing 210029, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China
| | - Xiaofei Liang
- Shaanxi Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji 721013, China
| | - Sheng Guo
- Nanjing University of Chinese Medicine/National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210029, China; Jiangsu Province Key Laboratory of High Technology Research, Nanjing 210029, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China
| | - Bing Qin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - E-Hu Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jin-Ao Duan
- Nanjing University of Chinese Medicine/National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210029, China; Jiangsu Province Key Laboratory of High Technology Research, Nanjing 210029, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China.
| |
Collapse
|
11
|
Wu H, Li Y, Shi L, Liu Y, Shen J. New Advances in Periodontal Functional Materials Based on Antibacterial, Anti-Inflammatory, and Tissue Regeneration Strategies. Adv Healthc Mater 2025; 14:e2403206. [PMID: 39895157 DOI: 10.1002/adhm.202403206] [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: 08/26/2024] [Revised: 01/17/2025] [Indexed: 02/04/2025]
Abstract
With the global population aging, awareness of oral health is rising. Periodontitis, a widespread bacterial infectious disease, is gaining attention. Current novel biomaterials address key clinical issues like bacterial infection, gum inflammation, tooth loosening, and loss, focusing on antibacterial, anti-inflammatory, and tissue regeneration properties. However, strategies that integrate the advantages of these biomaterials to achieve synergistic therapeutic effects by clearing oral biofilms, inhibiting inflammation activation, and restoring periodontal soft and hard tissue functions remain very limited. Recent studies highlight the link between periodontitis and systemic diseases, underscoring the complexity of the periodontal disease. There is an urgent need to find comprehensive treatment plans that address clinical requirements. Whether by integrating new biomaterials to enhance existing periodontal treatments or by developing novel approaches to replace traditional therapies, these efforts will drive advancements in periodontitis treatment. Therefore, this review compares novel biomaterials with traditional treatments. It highlights the design concepts and mechanisms of these functional materials, focusing on their antibacterial, anti-inflammatory, and tissue regeneration properties, and discusses the importance of developing comprehensive treatment strategies. This review aims to provide guidance for emerging periodontitis research and to promote the development of precise and efficient treatment strategies.
Collapse
Affiliation(s)
- Haoyue Wu
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yuanfeng Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jing Shen
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| |
Collapse
|
12
|
Zhu W, Yang Z, Zhou S, Zhang J, Xu Z, Xiong W, Liu P. Modic changes: From potential molecular mechanisms to future research directions (Review). Mol Med Rep 2025; 31:90. [PMID: 39918002 PMCID: PMC11836598 DOI: 10.3892/mmr.2025.13455] [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: 10/09/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
Low back pain (LBP) is a leading cause of disability worldwide. Although not all patients with Modic changes (MCs) experience LBP, MC is often closely associated with LBP and disc degeneration. In clinical practice, the focus is usually on symptoms related to MC, which are hypothesized to be associated with LBP; however, the link between MC and nerve compression remains unclear. In cases of intervertebral disc herniation, nerve compression is often the definitive cause of symptoms. Recent advances have shed light on the pathophysiology of MC, partially elucidating its underlying mechanisms. The pathogenesis of MC involves complex bone marrow‑disc interactions, resulting in bone marrow inflammation and edema. Over time, hematopoietic cells are gradually replaced by adipocytes, ultimately resulting in localized bone marrow sclerosis. This process creates a barrier between the intervertebral disc and the bone marrow, thereby enhancing the stability of the vertebral body. The latest understanding of the pathophysiology of MC suggests that chronic inflammation plays a significant role in its development and hypothesizes that the complement system may contribute to its pathological progression. However, this hypothesis requires further research to be confirmed. The present review we proposed a pathological model based on current research, encompassing the transition from Modic type 1 changes (MC1) to Modic type 2 changes (MC2). It discussed key cellular functions and their alterations in the pathogenesis of MC and outlined potential future research directions to further elucidate its mechanisms. Additionally, it reviewed the current clinical staging and pathogenesis of MC, recommended the development of an updated staging system and explored the prospects of integrating emerging artificial intelligence technologies.
Collapse
Affiliation(s)
- Weijian Zhu
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhou Yang
- Department of Orthopedics, Hongxin Harmony Hospital, Li Chuan, Hubei 445400 P.R. China
| | - Sirui Zhou
- Department of Respiration, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Jinming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhihao Xu
- Department of Hepatobiliary Surgery, Huaqiao Hospital, Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Wei Xiong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ping Liu
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| |
Collapse
|
13
|
Zhao F, Chen Y, Dong A, Song K. Experimental study of radial extracorporeal shock wave therapy for periprosthetic osteolysis induced by wear particles. J Orthop Surg Res 2025; 20:282. [PMID: 40083028 PMCID: PMC11908048 DOI: 10.1186/s13018-025-05661-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
Radial Extracorporeal Shock Wave Therapy (rESWT) is applied as a conservative treatment modality in orthopedics, yet its effectiveness in addressing aseptic loosening of cementless joint prostheses remains unclear. Through animal experimentation, we have revealed that in a titanium particle-induced osteolysis rat model, rESWT intervention significantly increased periprosthetic bone density compared to untreated controls, concurrently reducing osteolytic lesion area and lowering serum IL-1β levels. Histological analyses demonstrated a relative decrease in osteoclast counts within the treatment group versus non-treated controls. These findings indicate that rESWT, through mechanisms involving anti-inflammatory actions and suppression of osteoclastogenesis, may serve as a non-invasive therapeutic strategy for preventing and managing periprosthetic bone loss, demonstrating clinical potential to delay or eliminate the necessity for revision surgeries.
Collapse
Affiliation(s)
| | - Yufei Chen
- First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ao Dong
- First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Keguan Song
- First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
14
|
Ai H, Dou C, Wu Y, Zhang D, Zhang Z, Zhang C, Xi Y, Qu Y, Tan J, Yin P, Xu J, Guo S, Luo F. Osteoclast-derived apoptotic bodies accelerate the pathological progression of osteoarthritis via disturbing subchondral bone remodeling. J Orthop Translat 2025; 51:108-118. [PMID: 40123999 PMCID: PMC11930187 DOI: 10.1016/j.jot.2025.01.004] [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: 06/03/2024] [Revised: 12/03/2024] [Accepted: 01/10/2025] [Indexed: 03/25/2025] Open
Abstract
Objective To investigate the role of osteoclast-derived apoptotic bodies (OC-ABs) in osteoarthritis (OA), specifically their impact on subchondral bone remodeling and disease progression, and to explore potential therapeutic strategies targeting OC-AB-induced pathways. Methods We utilized a mouse model of anterior cruciate ligament transection (ACLT) to simulate post-traumatic osteoarthritis (PTOA). Levels of OC-ABs were assessed in subchondral bone and correlated with OA severity. Additionally, apoptotic body-deficient MRL/lpr mice were analyzed to evaluate the direct contribution of OC-ABs to OA progression and subchondral bone remodeling. The involvement of OC-ABs in osteogenesis was further examined using mesenchymal stem cells (MSCs), with a focus on the RANKL reverse signaling pathway. The therapeutic potential of rapamycin to counteract OC-AB effects was tested. Results Increased OC-AB accumulation in subchondral bone was positively correlated with OA severity in ACLT-induced mice. Apoptotic body-deficient MRL/lpr mice demonstrated slower OA progression and maintained more stable subchondral bone architecture, indicating a pathogenic role of OC-ABs in OA. OC-ABs significantly stimulated osteogenesis in MSCs via the RANKL reverse signaling pathway. Treatment with rapamycin effectively reversed OC-AB-induced subchondral bone formation, mitigated OA progression, and inhibited the RANKL reverse signaling pathway. Conclusion OC-ABs play a critical role in exacerbating OA by promoting subchondral bone remodeling via the RANKL reverse signaling pathway. Rapamycin presents as a promising therapeutic agent capable of mitigating OC-AB-driven pathology, highlighting new avenues for targeted OA treatment.
Collapse
Affiliation(s)
- Hongbo Ai
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ce Dou
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yutong Wu
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Department of Orthopedics, 75th Group Army Hospital, Dali, 671000, China
| | - Dongyang Zhang
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ziyang Zhang
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chao Zhang
- Department of Orthopedics, 75th Group Army Hospital, Dali, 671000, China
| | - Yuhang Xi
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ying Qu
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiulin Tan
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, 100853, China
| | - Jianzhong Xu
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shuquan Guo
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Fei Luo
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
15
|
Lee SH, Lee KH, Kim D, Jeon C, Whangbo M, Jo HR, Youn J, Lee CH, Choi SH, Park YS, Nam B, Jo S, Kim TH. Targeting osteoclast-derived DPP4 alleviates inflammation-mediated ectopic bone formation in ankylosing spondylitis. Arthritis Res Ther 2025; 27:40. [PMID: 40001226 PMCID: PMC11853818 DOI: 10.1186/s13075-025-03474-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/03/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Ankylosing spondylitis (AS) is a chronic inflammatory disease characterized by ectopic bone formation. The anti-inflammatory function of dipeptidyl peptidase-4 (DPP4) inhibitor has been reported in bone metabolism, but its utility in AS has not previously been investigated. METHODS We assessed DPP4 level in serum, synovial fluid, and facet joint tissue of AS patients. Additionally, we investigated the effect of a DPP4 inhibitor in an experimental AS model using curdlan-injected SKG mice. Following curdlan injection, SKG mice were orally administered a DPP4 inhibitor three times per week for 5 weeks and observed clinical arthritis scores, and analyzed by micro-CT. Furthermore, osteoclast precursor cells (OPCs) from curdlan-injected SKG mice were treated with DPP4 inhibitor and evaluated the inhibitory effects of this treatment in vitro. RESULTS Soluble DPP4 level was elevated in the serum and synovial fluid of patients with AS compared to those in the control group. Expression of DPP4 increased gradually during human osteoclastogenesis and was high in mature osteoclasts. Oral administration of a DPP4 inhibitor resulted in a decrease in thickness of the hind paw, clinical arthritis scores, and enthesitis at the ankle in curdlan-injected SKG mice compared to the vehicle group. Micro-CT data revealed a significant reduction in inflammation-induced low bone density in the DPP4 inhibitor group. Moreover, treatment with a DPP4 inhibitor significantly reduced osteoclast differentiation of OPC in addition to decreasing expression of osteoclast differentiation markers. CONCLUSION Our findings suggest that inhibiting DPP4 may have a therapeutic effect on inflammation-mediated ectopic bone formation in AS patients.
Collapse
Affiliation(s)
- Seung Hoon Lee
- Hanyang University Institute for Rheumatology Research (HYIRR), Hanyang University, Seoul, 04763, Korea
| | - Kyu Hoon Lee
- Department of Rehabilitation Medicine, Hanyang University Hospital for Rheumatic Diseases, Seoul, 04763, Korea
| | - Dongju Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Hanyang University, Seoul, 04763, Korea
| | - Chanhyeok Jeon
- Hanyang University Institute for Rheumatology Research (HYIRR), Hanyang University, Seoul, 04763, Korea
| | - Min Whangbo
- Hanyang University Institute for Rheumatology Research (HYIRR), Hanyang University, Seoul, 04763, Korea
| | - Hye-Ryeong Jo
- Hanyang University Institute for Rheumatology Research (HYIRR), Hanyang University, Seoul, 04763, Korea
| | - Jeehee Youn
- Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul, 04763, South Korea
| | - Chang-Hun Lee
- Department of Orthopaedic Surgery, Hanyang University Seoul Hospital, Seoul, 04763, South Korea
| | - Sung Hoon Choi
- Department of Orthopaedic Surgery, Hanyang University Seoul Hospital, Seoul, 04763, South Korea
| | - Ye-Soo Park
- Department of Orthopaedic Surgery, Guri Hospital, Hanyang University College of Medicine, Guri, 11923, South Korea
| | - Bora Nam
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Sungsin Jo
- Department of Biology, College of Natural Sciences, Soonchunhyang University, 22 Soonchunhyang-ro, Shinchang-myeon, Asan city, Chungcheongnam-do, 31538, Republic of Korea.
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Hanyang University, Seoul, 04763, Korea.
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea.
| |
Collapse
|
16
|
Suamphan S, Makeudom A, Krisanaprakornkit S, Meekhantong P, Dechtham E, Leethanakul C. Enhanced osteogenic differentiation of human periodontal ligament cells by mature osteoclasts. J Oral Biosci 2025; 67:100632. [PMID: 39993474 DOI: 10.1016/j.job.2025.100632] [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: 11/26/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
OBJECTIVE Several in vitro studies have shown that reverse signaling from osteoclasts regulates osteoblast differentiation and mineralization. However, none of these studies have reported the effects of this signaling pathway on periodontal ligament (PDL) cells. Therefore, in this study, we aimed to investigate the interaction between receptor activators of nuclear factor kappa B (RANK) released from mature human osteoclasts and the membranous RANK ligand (RANKL) in human PDL cells. METHODS Multinucleated mature human osteoclasts were differentiated from peripheral blood mononuclear cells upon incubation with recombinant macrophage colony-stimulating factor and RANKL. Mature osteoclasts and human PDL cells were characterized. A mature osteoclast-conditioned medium (OC-CM) was used to induce osteogenic differentiation of PDL cells. Mechanistic analysis of RANK-RANKL reverse signaling were conducted to determine the regulation of osteogenic induction using conditioned medium from mature osteoclasts treated with GW4869 (GW-OC-CM) or PDL cells pretreated with recombinant human osteoprotegerin (OPG). RESULTS OC-CM significantly upregulated the mRNA expression of osteogenic genes and enhanced the osteogenic differentiation and biomineralization of PDL cells (p < 0.05). GW-OC-CM significantly reduced the expression of osteogenic genes, osteogenic differentiation, and biomineralization in PDL cells (p < 0.05). Similarly, the pretreatment of PDL cells with OPG before OC-CM treatment significantly reduced the osteogenic induction of PDL cells (p < 0.05). CONCLUSION Mature osteoclasts can induce osteogenesis in human PDL cells via RANK-RANKL reverse signaling.
Collapse
Affiliation(s)
- Sumit Suamphan
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Anupong Makeudom
- School of Dentistry, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | | | | | - Ekapong Dechtham
- School of Dentistry, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Chidchanok Leethanakul
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Center of Excellence for Oral Health, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand.
| |
Collapse
|
17
|
Liu L, Chen H, Zhao X, Han Q, Xu Y, Liu Y, Zhang A, Li Y, Zhang W, Chen B, Wang J. Advances in the application and research of biomaterials in promoting bone repair and regeneration through immune modulation. Mater Today Bio 2025; 30:101410. [PMID: 39811613 PMCID: PMC11731593 DOI: 10.1016/j.mtbio.2024.101410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/02/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
With the ongoing development of osteoimmunology, increasing evidence indicates that the local immune microenvironment plays a critical role in various stages of bone formation. Consequently, modulating the immune inflammatory response triggered by biomaterials to foster a more favorable immune microenvironment for bone regeneration has emerged as a novel strategy in bone tissue engineering. This review first examines the roles of various immune cells in bone tissue injury and repair. Then, the contributions of different biomaterials, including metals, bioceramics, and polymers, in promoting osteogenesis through immune regulation, as well as their future development directions, are discussed. Finally, various design strategies, such as modifying the physicochemical properties of biomaterials and integrating bioactive substances, to optimize material design and create an immune environment conducive to bone formation, are explored. In summary, this review comprehensively covers strategies and approaches for promoting bone tissue regeneration through immune modulation. It offers a thorough understanding of current research trends in biomaterial-based immune regulation, serving as a theoretical reference for the further development and clinical application of biomaterials in bone tissue engineering.
Collapse
Affiliation(s)
- Li Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Hao Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Xue Zhao
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Qing Han
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Yongjun Xu
- Department of Orthopedics Surgery, Wangqing County People's Hospital, Yanbian, 133000, Jilin, China
| | - Yang Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Aobo Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Yongyue Li
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Weilong Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Bingpeng Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Jincheng Wang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| |
Collapse
|
18
|
Kitazawa S, Haraguchi R, Kitazawa R. Roles of osteoclasts in pathological conditions. Pathol Int 2025; 75:55-68. [PMID: 39704061 PMCID: PMC11849001 DOI: 10.1111/pin.13500] [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: 11/06/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024]
Abstract
Bone is a unique organ crucial for locomotion, mineral metabolism, and hematopoiesis. It maintains homeostasis through a balance between bone formation by osteoblasts and bone resorption by osteoclasts, which is regulated by the basic multicellular unit (BMU). Abnormal bone metabolism arises from an imbalance in the BMU. Osteoclasts, derived from the monocyte-macrophage lineage, are regulated by the RANKL-RANK-OPG system, which is a key factor in osteoclast differentiation. RANKL activates osteoclasts through its receptor RANK, while OPG acts as a decoy receptor that inhibits RANKL. In trabecular bone, high turnover involves rapid bone formation and resorption, influenced by conditions such as malignancy and inflammatory cytokines that increase RANKL expression. Cortical bone remodeling, regulated by aged osteocytes expressing RANKL, is less understood, despite ongoing research into how Rett syndrome, characterized by MeCP2 abnormalities, affects RANKL expression. Balancing trabecular and cortical bone involves mechanisms that preserve cortical bone, despite overall bone mass reduction due to aging or oxidative stress. Research into genes like sFRP4, which modulates bone mass, highlights the complex regulation by BMUs. The roles of the RANKL-RANK-OPG system extend beyond bone, affecting processes such as aortic valve formation and temperature regulation, which highlight the interconnected nature of biological research.
Collapse
Affiliation(s)
- Sohei Kitazawa
- Department of Molecular PathologyEhime University Graduate School of Medicine, ShitsukawaToon CityJapan
| | - Ryuma Haraguchi
- Department of Molecular PathologyEhime University Graduate School of Medicine, ShitsukawaToon CityJapan
| | - Riko Kitazawa
- Department of Molecular PathologyEhime University Graduate School of Medicine, ShitsukawaToon CityJapan
- Division of Diagnostic PathologyEhime University Hospital, ShitsukawaToon CityJapan
| |
Collapse
|
19
|
Feng Y, Mo Y, Zhang Y, Teng Y, Xi D, Zhou J, Zeng G, Zong S. Polyphyllin VI: A promising treatment for prostate cancer bone metastasis. Int Immunopharmacol 2025; 144:113684. [PMID: 39602960 DOI: 10.1016/j.intimp.2024.113684] [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: 01/30/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Prostate cancer, as one of the most prevalent malignant tumors in men, seriously affects the prognosis and survival of patients due to its extremely high rate of bone metastasis. This study investigated the effect of Polyphyllin VI (PPVI) on metastatic bone disease for the first time in prostate cancer, focusing on its impact on osteoclast and tumor cell. In vitro studies utilized TRAP staining, ghost pen cyclic peptide staining, and bone resorption assays to evaluate the differentiation and function of receptor activator of nuclear factor-κB ligand (RANKL) induced and RM-1 conditional medium (CM) induced osteoclasts. The colony formation assay, wound healing assay, and Transwell assay were employed to analyze tumor cell proliferation, migration, and invasion in vitro. Flow cytometry was used to detect the cycling and apoptosis of tumor cells in vitro. Western Blot and PCR assays were conducted to assess the expression of genes. In vivo, micro-CT, hematoxylin-eosin staining, and immunohistochemical staining evaluated the impact of PPVI on bone destruction and tumor growth in a mouse model of tumor tibial metastasis. The study results indicated that PPVI effectively inhibited osteoclast differentiation, suppresses tumor cell proliferation, migration, and invasion in vitro, and induces apoptosis and G2/M phase arrest. In vivo, PPVI not only inhibits the growth of metastatic tumors but also mitigates the resulting bone destruction. These results suggest that PPVI holds significant potential as an alternative treatment for prostate cancer with bone metastasis, providing insights into its molecular mechanisms and therapeutic efficacy.
Collapse
Affiliation(s)
- Yanbin Feng
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Jinan, Shandong, China
| | - Yaomin Mo
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Yang Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Jinan, Shandong, China
| | - Yilin Teng
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Deshuang Xi
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Junhong Zhou
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Gaofeng Zeng
- Department of Nutrition and Food Hygiene, College of Public Hygiene of Guangxi Medical University, Nanning, Guangxi, China.
| | - Shaohui Zong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
20
|
Zhang C, Yu M, Zhang L, Zhou X, Han J, Fu B, Xue H, Zhang C. Exploring the Analgesic Effect of Acupuncture on Knee Osteoarthritis Based on MLT/cAMP/PKA/CREB Signaling Pathway. J Inflamm Res 2025; 18:237-249. [PMID: 39802514 PMCID: PMC11724624 DOI: 10.2147/jir.s498202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
Background Acupuncture is an effective treatment for knee osteoarthritis (KOA), reducing pain and improving function. While melatonin (MLT) has notable pain relief benefits, the analgesic mechanism of acupuncture in KOA and its relationship with melatonin are still unknown. This study aims to explore this mechanism. Methods In this work, the KOA rabbit model was constructed using the traditional Hulth method, and the therapeutic effect was assessed by the Lequesne MG score and Pain assessment by hot plate test. The pathological alterations of cartilage tissue were observed using hematoxylin and eosin (H&E) staining, Safranin O-fast green and MASSON staining to observe the pathological changes in cartilage tissue, and the efficacy was evaluated according to the principles of Mankin score and Osteoarthritis Research Society International (OARSI) score. Meanwhile, MLT in serum, cyclic adenosine monophosphate (cAMP) in cartilage, and matrix metalloproteinase-3 (MMP-3) in joint fluid were detected by enzyme-linked immunosorbent assay. In addition, the expression of aromatic L-amino acid N-acetyltransferase (AANAT), melatonin receptor 1 (MT1) and 2 (MT2) mRNAs in cartilage was determined by real-time quantitative reverse transcription-polymerase chain reaction, and the levels of proteins related to PKA/CREB signaling pathway were detected by Western blotting. Results Based on the results of Lequesne MG score and Pain assessment by hot plate test experimental data, the treatment group presented significant improvements in knee pain and overall function relative to OA (Osteoarthritis) group. Besides, according to results of histologic staining, Mankin and OARSI scores, articular cartilage degeneration of treatment group remarkably improved. In addition, acupuncture significantly reduced the expression of the inflammatory factor MMP-3 in knee joint fluid and significantly increased the levels of MLT, AANAT, MT1, MT2, cAMP, PKA and CREB. Conclusion By regulating sympathetic excitability, acupuncture may activate the MLT/cAMP/PKA/CREB signaling pathway, decrease inflammatory factor expression and slow down degradation of articular cartilage, resulting in the relief of knee pain.
Collapse
Affiliation(s)
- Chao Zhang
- Orthopedics Department, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, People’s Republic of China
| | - Man Yu
- Department of Nephrology and Rheumatology, Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300250, People’s Republic of China
| | - Longyao Zhang
- Orthopedics Department, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, People’s Republic of China
| | - Xin Zhou
- Orthopedics Department, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, People’s Republic of China
| | - Jinchang Han
- Orthopedics Department, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, People’s Republic of China
| | - Bifeng Fu
- Orthopedics Department, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, People’s Republic of China
| | - Hongfei Xue
- Orthopedics Department, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, People’s Republic of China
| | - Chao Zhang
- Orthopedics Department, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, People’s Republic of China
| |
Collapse
|
21
|
Luo H, Lin S, Lv H, Tan W, Zhong J, Xiong J, Liu Z, Wu Q, Chen M, Cao K. Chrysoeriol: a natural RANKL inhibitor targeting osteoclastogenesis and ROS regulation for osteoporosis therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03714-3. [PMID: 39755833 DOI: 10.1007/s00210-024-03714-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/07/2024] [Indexed: 01/06/2025]
Abstract
Chrysoeriol (CHE) is a naturally occurring compound with established anti-inflammatory and anti-tumor effects. This study examines its potential role in regulating osteoclast differentiation and activity, both of which are crucial for bone remodeling. Computational docking revealed high binding affinity between CHE and RANKL, specifically at the Lys-181 residue of RANKL, suggesting potential inhibitory interactions on osteoclastogenesis. In vitro assays confirmed CHE's non-toxic profile at concentrations below 20 μM and demonstrated a dose-dependent suppression of osteoclast differentiation. Notably, CHE treatment significantly reduced TRAP activity and bone resorption capacity in a dose-dependent manner. Furthermore, CHE markedly decreased ROS production by NOX-1 expression and modulated the NRF2/KEAP1 pathway to enhance ROS clearance. The compound also showed inhibitory effects on the NF-κB and MAPK signaling pathways, which are crucial for osteoclast activation. In an ovariectomized mouse model, administration of CHE mitigated bone loss, indicating its therapeutic potential in osteoporosis. Collectively, these findings establish CHE as a promising natural therapeutic agent for treating bone disorders characterized by excessive bone resorption, underscoring the need for further clinical investigation.
Collapse
Affiliation(s)
- Hao Luo
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Sijian Lin
- The Rehabilitation Medicine Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Hao Lv
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Wen Tan
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Junlong Zhong
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Jiachao Xiong
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - ZhiMing Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Qin Wu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Ming Chen
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Kai Cao
- The Key Laboratory of Spine and Spinal Cord Disease of Jiangxi Province, Nanchang, 330006, China.
- Department of Orthopedics, Affiliated Rehabilitation Hospital of Nanchang University, Nanchang, 330002, China.
| |
Collapse
|
22
|
Fu YF, Shi SW, Wu JJ, Yuan ZD, Wang LS, Nie H, Zhang ZY, Wu X, Chen YC, Ti HB, Zhang KY, Mao D, Ye JX, Li X, Yuan FL. Osteoclast Secretes Stage-Specific Key Molecules for Modulating Osteoclast-Osteoblast Communication. J Cell Physiol 2025; 240:e31484. [PMID: 39606839 DOI: 10.1002/jcp.31484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
In most cases of bone metabolic disorders, such as osteoporosis and osteomalacia, these conditions are often attributed to dysfunctional osteoclasts, leading to their common characterization as "destructors." In addition to the widely documented regulatory process where osteoblasts direct osteoclastic bone resorption, there is increasing evidence suggesting that osteoclasts also in turn influence osteoblastic bone formation through direct and indirect mechanisms. It is well-known that differentiation of osteoclasts involves several stages, each characterized by specific cellular features and functions. Stage-specific key molecules secreted during these stages play a critical role in mediating osteoclast-osteoblast communication. In this review, we described the different stages of osteoclast differentiation and reviewed stage-specific key molecules involved in osteoclasts-osteoblasts communication. We highlighted that a detailed understanding of these processes and molecular mechanism could facilitate the development of novel treatments for bone metabolic disorders.
Collapse
Affiliation(s)
- Yi-Fei Fu
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Shu-Wen Shi
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
| | - Zheng-Dong Yuan
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
| | - Lei-Sheng Wang
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Hao Nie
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
| | - Zheng-Yu Zhang
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Xian Wu
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Yue-Chun Chen
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Hui-Bo Ti
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Ke-Yue Zhang
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Dong Mao
- Orthopaedic Institute, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, China
| | - Jun-Xing Ye
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Xia Li
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| |
Collapse
|
23
|
Sobacchi C, Menale C, Crisafulli L, Ficara F. Role of RANKL Signaling in Bone Homeostasis. Physiology (Bethesda) 2025; 40:0. [PMID: 39255276 DOI: 10.1152/physiol.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
RANKL and its cognate receptor RANK are crucial regulators of bone metabolism in physiological as well as in pathological conditions. Here we go through the works that unveiled the paramount role of this signaling pathway. We focus on the RANKL cytokine, whose alterations are responsible for rare and common bone diseases. We describe recent insights on the regulation of RANKL expression, which provide new hints for the pharmacological regulation of this molecule. Based on the multiple functions exerted by RANKL (within and outside the bone tissue), we advise caution regarding the potential unintended consequences of its inhibition.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| | - Ciro Menale
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," Naples, Italy
| | - Laura Crisafulli
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| | - Francesca Ficara
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
24
|
Pérez-Chacón G, Santamaría PG, Redondo-Pedraza J, González-Suárez E. RANK/RANKL Signaling Pathway in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:309-345. [PMID: 39821032 DOI: 10.1007/978-3-031-70875-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
RANK pathway has attracted increasing interest as a promising target in breast cancer, given the availability of denosumab, an anti-RANKL drug. RANK signaling mediates progesterone-driven regulation of mammary gland development and favors breast cancer initiation by controlling mammary cell proliferation and stem cell fate. RANK activation promotes luminal mammary epithelial cell senescence, acting as an initial barrier to tumorigenesis but ultimately facilitating tumor progression and metastasis. Comprehensive analyses have demonstrated that RANK protein expression is an independent biomarker of poor prognosis in postmenopausal and estrogen receptor-negative breast cancer patients. RANK pathway also has multiple roles in immunity and inflammation, regulating innate and adaptive responses. In the tumor microenvironment, RANK and RANKL are expressed by different immune cell populations and contribute to the regulation of tumor immune surveillance, mainly driving immunosuppressive effects.Herein, we discuss the preventive and therapeutic potential of targeting RANK signaling in breast cancer given its tumor cell intrinsic and extrinsic effects. RANKL inhibition has been shown to induce mammary tumor cell differentiation and an antitumor immune response. Moreover, loss of RANK signaling increases sensitivity of breast cancer cells to chemotherapy, targeted therapies such as HER2 and CDK4/6 inhibitors, and immunotherapy. Finally, we describe clinical trials of denosumab for breast cancer prevention, such as those ongoing in women with high risk of developing breast cancer, large phase III clinical trials where the impact of adjuvant denosumab on disease-free survival has been assessed, and window trials to evaluate the immunomodulatory effects of denosumab in breast cancer and other solid tumors.
Collapse
Affiliation(s)
- Gema Pérez-Chacón
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | - Eva González-Suárez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
25
|
Yang XD, Haga CL, Phinney DG. Signaling Dynamics in Osteogenesis: Unraveling Therapeutic Targets for Bone Generation. Curr Drug Targets 2025; 26:350-366. [PMID: 39791147 DOI: 10.2174/0113894501359782241216082049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025]
Abstract
Diseases affecting bone encompass a spectrum of disorders, from prevalent conditions such as osteoporosis and Paget's disease, collectively impacting millions, to rare genetic disorders including Fibrodysplasia Ossificans Progressiva (FOP). While several classes of drugs, such as bisphosphonates, synthetic hormones, and antibodies, are utilized in the treatment of bone diseases, their efficacy is often curtailed by issues of tolerability and high incidence of adverse effects. Developing therapeutic agents for bone diseases is hampered by the fact that numerous pathways regulating bone metabolism also perform pivotal functions in other organ systems. Consequently, the selection of an appropriate target is a complicated process despite the significant demand for novel medications to address bone diseases. Research has shown the role of various cell signaling pathways, including Wnt, PTHR1, CASR, BMPRs, OSCAR, and TWIST1, in the regulation of osteogenesis, bone remodeling, and homeostasis. Disruptions in bone homeostasis can result in decreased bone density and the onset of osteoporosis. There remains a need for the development of drugs that can enhance bone remodeling with improved side effects profiles. The exploration of promising targets to stimulate bone formation has the potential to significantly advance the field of bone-related medical care, thereby improving the quality of life for millions. Additionally, a deeper understanding of anabolic and catabolic pathway mechanisms could enable future studies to explore synergistic effects between unrelated pathways. Herein, we explore potential drug targets that may be exploited therapeutically using small molecule agonists or antagonists to promote bone remodeling and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Xue D Yang
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL33458, USA
| | - Christopher L Haga
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL33458, USA
| | - Donald G Phinney
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL33458, USA
| |
Collapse
|
26
|
Chang W, Tian B, Qin Q, Li D, Zhang Y, Zhou C, Wu B, Zhang M, Shan H, Ni Y, Dong Q, Wang C, Zhou XZ, Bai J. Receptor Activator of Nuclear Factor Kappa-B-Expressing Mesenchymal Stem Cells-Derived Extracellular Vesicles for Osteoporosis Therapy. ACS NANO 2024; 18:35368-35382. [PMID: 39692894 DOI: 10.1021/acsnano.4c12064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
The dynamic balance between bone resorption and formation is critical for maintaining healthy bone homeostasis. However, the receptor activator of the nuclear factor B ligand (RANKL) primarily stimulates mature osteoclasts to resorb bone, and its upregulation leads to osteoporosis in patients. Here, we designed RANK-expressing extracellular vesicles (EVs) derived from mesenchymal stem cells to maintain bone homeostasis in mice. This engineered EV (EV@R) effectively neutralizes excess RANKL in bone tissue due to the RANK-RANKL interaction, thereby attenuating osteoclast differentiation. Additionally, we found that miRNA-21a-5p in EV@R contributes to restoring bone metabolic homeostasis. We demonstrate the protective and therapeutic efficacy of EV@R against osteoporosis in the ovariectomy-induced osteoporosis mouse model with a lasting effect and minimal side effects. Our study provides an alternative way to use engineered EVs for bone homeostasis treatment.
Collapse
Affiliation(s)
- Wenju Chang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
- Anhui Province Key Laboratory of Tissue Transplantation (Bengbu Medical University), Bengbu, Anhui 233004, China
| | - Bo Tian
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Qin Qin
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Dongxiao Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Yue Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chenmeng Zhou
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bingbing Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Mingchao Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Huajian Shan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Yichao Ni
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Qirong Dong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xiao-Zhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Jinyu Bai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| |
Collapse
|
27
|
Yin Y, Tang Q, Yang J, Gui S, Zhang Y, Shen Y, Zhou X, Yu S, Chen G, Sun J, Han Z, Zhang L, Chen L. Endothelial BMAL1 decline during aging leads to bone loss by destabilizing extracellular fibrillin-1. J Clin Invest 2024; 134:e176660. [PMID: 39680455 DOI: 10.1172/jci176660] [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: 10/13/2023] [Accepted: 10/15/2024] [Indexed: 12/18/2024] Open
Abstract
The occurrence of aging is intricately associated with alterations in circadian rhythms that coincide with stem cell exhaustion. Nonetheless, the extent to which the circadian system governs skeletal aging remains inadequately understood. Here, we noticed that skeletal aging in male mice was accompanied by a decline in a core circadian protein, BMAL1, especially in bone marrow endothelial cells (ECs). Using male mice with endothelial KO of aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1), we ascertained that endothelial BMAL1 in bone played a crucial role in ensuring the stability of an extracellular structural component, fibrillin-1 (FBN1), through regulation of the equilibrium between the extracellular matrix (ECM) proteases thrombospondin type 1 domain-containing protein 4 (THSD4) and metalloproteinase with thrombospondin motifs 4 (ADAMTS4), which promote FBN1 assembly and breakdown, respectively. The decline of endothelial BMAL1 during aging prompted excessive breakdown of FBN1, leading to persistent activation of TGF-β/SMAD3 signaling and exhaustion of bone marrow mesenchymal stem cells. Meanwhile, the free TGF-β could promote osteoclast formation. Further analysis revealed that activation of ADAMTS4 in ECs lacking BMAL1 was stimulated by TGF-β/SMAD3 signaling through an ECM-positive feedback mechanism, whereas THSD4 was under direct transcriptional control by endothelial BMAL1. Our investigation has elucidated the etiology of bone aging in male mice by defining the role of ECs in upholding the equilibrium within the ECM, consequently coordinating osteogenic and osteoclastic activities and retarding skeletal aging.
Collapse
Affiliation(s)
- Ying Yin
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Qingming Tang
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Jingxi Yang
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Shiqi Gui
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yifan Zhang
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yufeng Shen
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xin Zhou
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Shaoling Yu
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Guangjin Chen
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Jiwei Sun
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Zhenshuo Han
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Chen
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| |
Collapse
|
28
|
Freeman C, A S MD, A S P. Unraveling the Intricacies of OPG/RANKL/RANK Biology and Its Implications in Neurological Disorders-A Comprehensive Literature Review. Mol Neurobiol 2024; 61:10656-10670. [PMID: 38777981 DOI: 10.1007/s12035-024-04227-z] [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: 02/03/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
The OPG/RANKL/RANK framework, along with its specific receptors, plays a crucial role in bone remodeling and the functioning of the central nervous system (CNS) and associated disorders. Recent research and investigations provide evidence that the components of osteoprotegerin (OPG), receptor activator of NF-kB ligand (RANKL), and receptor activator of NF-kB (RANK) are expressed in the CNS. The CNS structure encompasses cells involved in neuroinflammation, including local macrophages, inflammatory cells, and microglia that cross the blood-brain barrier. The OPG/RANKL/RANK trio modulates the neuroinflammatory response based on the molecular context. The levels of OPG/RANKL/RANK components can serve as biomarkers in the blood and cerebrospinal fluid. They act as neuroprotectants following brain injuries and also participate in the regulation of body weight, internal body temperature, brain ischemia, autoimmune encephalopathy, and energy metabolism. Although the OPG/RANKL/RANK system is primarily known for its role in bone remodeling, further exploring deeper into its multifunctional nature can uncover new functions and novel drug targets for diseases not previously associated with OPG/RANKL/RANK signaling.
Collapse
Affiliation(s)
- Chrisanne Freeman
- Department of Biotechnology, Bishop Heber College, Tamil Nadu, Tiruchirappalli, 620017, India.
| | - Merlyn Diana A S
- Department of Biotechnology, Bishop Heber College, Tamil Nadu, Tiruchirappalli, 620017, India
- Department of Zoology and Research Centre, Lady Doak College, Tamil Nadu, Madurai, 625002, India
| | - Priscilla A S
- Department of Zoology and Research Centre, Lady Doak College, Tamil Nadu, Madurai, 625002, India
| |
Collapse
|
29
|
Gostage J, Kostenuik P, Goljanek-Whysall K, Bellantuono I, McCloskey E, Bonnet N. Extra-osseous Roles of the RANK-RANKL-OPG Axis with a Focus on Skeletal Muscle. Curr Osteoporos Rep 2024; 22:632-650. [PMID: 39325366 PMCID: PMC11499344 DOI: 10.1007/s11914-024-00890-2] [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] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
PURPOSE OF REVIEW This review aims to consolidate recent observations regarding extra-osseous roles of the RANK-RANKL-OPG axis, primarily within skeletal muscle. RECENT FINDINGS Preclinical efforts to decipher a common signalling pathway that links the synchronous decline in bone and muscle health in ageing and disease disclosed a potential role of the RANK-RANKL-OPG axis in skeletal muscle. Evidence suggests RANKL inhibition benefits skeletal muscle function, mass, fibre-type switching, calcium homeostasis and reduces fall incidence. However, there still exists ambiguity regarding the exact mechanistic actions and subsequent functional improvements. Other potential RANK-RANKL-OPG extra-osseous roles include regulation of neural-inflammation and glucose metabolism. Growing evidence suggests the RANK-RANKL-OPG axis may play a regulatory role in extra-osseous tissues, especially in skeletal muscle. Targeting RANKL may be a novel therapy in ameliorating loss of muscle mass and function. More research is warranted to determine the causality of the RANK-RANKL-OPG axis in extra-osseous tissues, especially those affected by aging.
Collapse
Affiliation(s)
- John Gostage
- The Medical Research Council/Versus Arthritis Centre for Integrated Research Into Musculoskeletal Aging, CIMA, University of Liverpool, Liverpool, UK
- Division of Clinical Medicine, School of Medicine and Population Health, Healthy Lifespan Institute and the Centre for Integrated Research in Musculoskeletal Aging, University of Sheffield, Sheffield, UK
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland
| | - Paul Kostenuik
- School of Dentistry and Phylon Pharma Services, University of Michigan, Thousand Oaks, CA, USA
| | - Katarzyna Goljanek-Whysall
- The Medical Research Council/Versus Arthritis Centre for Integrated Research Into Musculoskeletal Aging, CIMA, University of Liverpool, Liverpool, UK
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland
| | - Ilaria Bellantuono
- The Medical Research Council/Versus Arthritis Centre for Integrated Research Into Musculoskeletal Aging, CIMA, University of Liverpool, Liverpool, UK
- Division of Clinical Medicine, School of Medicine and Population Health, Healthy Lifespan Institute and the Centre for Integrated Research in Musculoskeletal Aging, University of Sheffield, Sheffield, UK
| | - Eugene McCloskey
- The Medical Research Council/Versus Arthritis Centre for Integrated Research Into Musculoskeletal Aging, CIMA, University of Liverpool, Liverpool, UK
- Division of Clinical Medicine, School of Medicine and Population Health, Healthy Lifespan Institute and the Centre for Integrated Research in Musculoskeletal Aging, University of Sheffield, Sheffield, UK
| | - Nicolas Bonnet
- Service of Bone Diseases, Department of Medicine, Geneva University Hospital and Faculty of Medicine, Geneva, Switzerland.
| |
Collapse
|
30
|
Chen L, Zhang Z, Liu X. Role and Mechanism of Mechanical Load in the Homeostasis of the Subchondral Bone in Knee Osteoarthritis: A Comprehensive Review. J Inflamm Res 2024; 17:9359-9378. [PMID: 39600681 PMCID: PMC11590007 DOI: 10.2147/jir.s492415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Osteoarthritis (OA) is one of the most prevalent degenerative joint diseases, and the knee joint is particularly susceptible to it. It typically affects the entire joint and is marked by the erosion of cartilage integrity, chondrocytopenia, subchondral bone sclerosis and the mild synovial inflammation. Pathological changes in the subchondral bone often serve as initiating factors for joint degeneration. Various predisposing factors, including metabolic disorders, oxidative stress, and abnormal mechanical loading, regulate OA pathogenesis. Of them, mechanical loading is closely associated with the maintenance of the subchondral bone. Disrupted mechanical loading, leading to subchondral bone remodeling, can potentially trigger OA, whereas appropriate loading might ameliorate its progression. Therefore, this narrative review aimed to discuss existing knowledge and explore how mechanical loading mediates changes in the subchondral bone, influencing the development of knee osteoarthritis. Special emphasis is placed on its role and underlying mechanisms in maintaining joint homeostasis.
Collapse
Affiliation(s)
- Lin Chen
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Zhan Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Xueyong Liu
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
31
|
Du Y, Chen H, Zhou L, Guo Q, Gong S, Feng S, Guan Q, Shi P, Lv T, Guo Y, Yang C, Sun P, Li K, Xu S, Li L. REGγ is essential to maintain bone homeostasis by degrading TRAF6, preventing osteoporosis. Proc Natl Acad Sci U S A 2024; 121:e2405265121. [PMID: 39536082 PMCID: PMC11588133 DOI: 10.1073/pnas.2405265121] [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: 03/13/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Primary osteoporosis, manifesting as decreased bone mass and increased bone fragility, is a "silent disease" that is often ignored until a bone breaks. Accordingly, it is urgent to develop reliable biomarkers and novel therapeutic strategies for osteoporosis treatment. Here, we identified REGγ as a potential biomarker of osteoporotic populations through proteomics analysis. Next, we demonstrated that REGγ deficiency increased osteoclast activity and triggered bone mass loss in REGγ knockout (KO) and bone marrow-derive macrophage (BMM)-conditional REGγ KO mice. However, the osteoclast activity decreased in BMM-conditional REGγ overexpression mice. Mechanistically, we defined that REGγ-20S proteasome directly degraded TRAF6 to inhibit bone absorption in a ubiquitin-independent pathway. More importantly, BMM-conditional Traf6 KO with REGγ KO mice could "rescue" the osteoporosis phenotypes. Based on NIP30 (a REGγ "inhibitor") dephosphorylation by CKII inhibition activated the ubiquitin-independent degradation of TRAF6, we selected TTP22, an inhibitor of CKII, and defined that TTP22 could alleviate osteoporosis in vitro and in vivo. Overall, our study reveals a unique function of NIP30/REGγ/TRAF6 axis in osteoporosis and provides a potential therapeutic drug TTP22 for osteoporosis.
Collapse
Affiliation(s)
- Yingying Du
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Hui Chen
- Department of Trauma-Emergency and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai200240, China
- Joint Center for Translational Medicine, Shanghai Fifth People’s Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai200241, China
- School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Lei Zhou
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200080, China
| | - Qunfeng Guo
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai200003, China
| | - Shuangming Gong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Siyuan Feng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Qiujing Guan
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Peilin Shi
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Tongxin Lv
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Yilan Guo
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai200241, China
| | - Cheng Yang
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai200003, China
| | - Peng Sun
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai200241, China
| | - Kun Li
- Health Science Center, East China Normal University, Shanghai200241, China
| | - Shuogui Xu
- Department of Emergency and Trauma, The First Affiliated Hospital of Naval Medical University, Shanghai200433, China
| | - Lei Li
- Joint Center for Translational Medicine, Shanghai Fifth People’s Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai200241, China
- School of Life Sciences, East China Normal University, Shanghai200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing401120, China
- East China Normal University, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai200241, China
| |
Collapse
|
32
|
Mehrvar A, Akbari M, Khosroshahi EM, Nekavand M, Mokhtari K, Baniasadi M, Aghababaian M, Karimi M, Amiri S, Moazen A, Maghsoudloo M, Alimohammadi M, Rahimzadeh P, Farahani N, Vaghar ME, Entezari M, Hashemi M. The impact of exosomes on bone health: A focus on osteoporosis. Pathol Res Pract 2024; 263:155618. [PMID: 39362132 DOI: 10.1016/j.prp.2024.155618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Osteoporosis is a widespread chronic condition. Although standard treatments are generally effective, they are frequently constrained by side effects and the risk of developing drug resistance. A promising area of research is the investigation of extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, which play a crucial role in bone metabolism. Exosomes, in particular, have shown significant potential in both the diagnosis and treatment of osteoporosis. EVs derived from osteoclasts, osteoblasts, mesenchymal stem cells, and other sources can influence bone metabolism, while exosomes from inflammatory and tumor cells may exacerbate bone loss, highlighting their dual role in osteoporosis pathology. This review offers a comprehensive overview of EV biogenesis, composition, and function in osteoporosis, focusing on their diagnostic and therapeutic potential. We examine the roles of various types of EVs and their cargo-proteins, RNAs, and lipids-in bone metabolism. Additionally, we explore the emerging applications of EVs as biomarkers and therapeutic agents, emphasizing the need for further research to address current challenges and enhance EV-based strategies for managing osteoporosis.
Collapse
Affiliation(s)
- Amir Mehrvar
- Assistant Professor, Department of Orthopedics, Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadarian Akbari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrandokht Nekavand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Midwifery, Faculty of nursing and midwifery, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mojtaba Baniasadi
- Department of Orthopedic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran; MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Aghababaian
- Department of Orthopedic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansour Karimi
- MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- MD, Assistant Professor of Orthopaedic Surgery, Shohadaye Haftom-e-Tir Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Moazen
- Department of Orthopedics, Bone and Joint Reconstruction Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohammad Eslami Vaghar
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of gynecology, Faculty of Medicine, Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
33
|
Zhao Z, Wu C, Huangfu Y, Zhang Y, Zhang J, Huang P, Dong A, Wang Y, Deng J, Wang W, Feng Z. Bioinspired Glycopeptide Hydrogel Reestablishing Bone Homeostasis through Mediating Osteoclasts and Osteogenesis in Periodontitis Treatment. ACS NANO 2024; 18:29507-29521. [PMID: 39401162 DOI: 10.1021/acsnano.4c05677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Irreversible alveolar bone resorption is one of the important clinical manifestations of periodontitis, which is initiated by a plaque biofilm and exacerbated by the imbalance of osteoclast activity and osteogenesis, affecting a patient's masticatory function and resulting in a high recurrence rate of periodontitis. Herein, to reestablish bone homeostasis in periodontitis, a minocycline hydrochloride (MH)-loaded glycopeptide hydrogel (MH/GRWgel) is fabricated to mediate alveolar bone absorption through sequential antibacterial and RANKL-blocking activities. GRWgel shows an ECM-like fibrous and porous microstructure serving as a scaffold for cell proliferation and differentiation and holds the merits including injectability, self-healing properties, and good biocompatibility. After injection in situ, MH is released rapidly from the hydrogel in the early stage, demonstrating a potent antimicrobial effect to combat the biofilm in the deep periodontal pocket. Moreover, MH/GRWgel exhibits a high specific binding efficiency with RANKL to suppress osteoclast maturation by shielding the RANKL/RANK interaction and enhancing osteogenic differentiation, thereby synergistically regulating bone homeostasis. In the rat periodontitis model, MH/GRWgel significantly curtails periodontitis progression through antimicrobial activity, inhibition of alveolar bone resorption, and promotion of bone regeneration, which is superior to the treatment of a commercial gel. These findings suggest that MH/GRWgel with superiority in regulating bone homeostasis provides a promising therapeutic strategy for periodontitis.
Collapse
Affiliation(s)
- Zhezhe Zhao
- Department of Periodontology, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Chenxuan Wu
- Department of Periodontology, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Yini Huangfu
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Yufeng Zhang
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Ju Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| | - Pingsheng Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| | - Anjie Dong
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Yonglan Wang
- Department of Periodontology, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Jiayin Deng
- Department of Periodontology, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Weiwei Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| | - Zujian Feng
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| |
Collapse
|
34
|
Singh M, Singh P, Singh B, Sharma K, Kumar N, Singh D, Mastana S. Molecular Signaling Pathways and MicroRNAs in Bone Remodeling: A Narrative Review. Diseases 2024; 12:252. [PMID: 39452495 PMCID: PMC11507001 DOI: 10.3390/diseases12100252] [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: 08/03/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
Bone remodeling is an intricate process executed throughout one's whole life via the cross-talk of several cellular events, progenitor cells and signaling pathways. It is an imperative mechanism for regaining bone loss, recovering damaged tissue and repairing fractures. To achieve this, molecular signaling pathways play a central role in regulating pathological and causal mechanisms in different diseases. Similarly, microRNAs (miRNAs) have shown promising results in disease management by mediating mRNA targeted gene expression and post-transcriptional gene function. However, the role and relevance of these miRNAs in signaling processes, which regulate the delicate balance between bone formation and bone resorption, are unclear. This review aims to summarize current knowledge of bone remodeling from two perspectives: firstly, we outline the modus operandi of five major molecular signaling pathways, i.e.,the receptor activator of nuclear factor kappa-B (RANK)-osteoprotegrin (OPG) and RANK ligand (RANK-OPG-RANKL), macrophage colony-stimulating factor (M-CSF), Wnt/β-catenin, Jagged/Notch and bone morphogenetic protein (BMP) pathways in regards to bone cell formation and function; and secondly, the miRNAs that participate in these pathways are introduced. Probing the miRNA-mediated regulation of these pathways may help in preparing the foundation for developing targeted strategies in bone remodeling, repair and regeneration.
Collapse
Affiliation(s)
- Monica Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Puneetpal Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Baani Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Kirti Sharma
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Nitin Kumar
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Deepinder Singh
- VardhmanMahavir Health Care, Urban Estate Ph-II, Patiala 147002, India;
| | - Sarabjit Mastana
- Human Genomics Laboratory, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
| |
Collapse
|
35
|
Xiang Q, Li L, Ji W, Gawlitta D, Walboomers XF, van den Beucken JJJP. Beyond resorption: osteoclasts as drivers of bone formation. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:22. [PMID: 39392536 PMCID: PMC11469995 DOI: 10.1186/s13619-024-00205-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Emerging evidence illustrates that osteoclasts (OCs) play diverse roles beyond bone resorption, contributing significantly to bone formation and regeneration. Despite this, OCs remain mysterious cells, with aspects of their lifespan-from origin, fusion, alterations in cellular characteristics, to functions-remaining incompletely understood. Recent studies have identified that embryonic osteoclastogenesis is primarily driven by osteoclast precursors (OCPs) derived from erythromyeloid progenitors (EMPs). These precursor cells subsequently fuse into OCs essential for normal bone development and repair. Postnatally, hematopoietic stem cells (HSCs) become the primary source of OCs, gradually replacing EMP-derived OCs and assuming functional roles in adulthood. The absence of OCs during bone development results in bone structure malformation, including abnormal bone marrow cavity formation and shorter long bones. Additionally, OCs are reported to have intimate interactions with blood vessels, influencing bone formation and repair through angiogenesis regulation. Upon biomaterial implantation, activation of the innate immune system ensues immediately. OCs, originating from macrophages, closely interact with the immune system. Furthermore, evidence from material-induced bone formation events suggests that OCs are pivotal in these de novo bone formation processes. Nevertheless, achieving a pure OC culture remains challenging, and interpreting OC functions in vivo faces difficulties due to the presence of other multinucleated cells around bone-forming biomaterials. We here describe the fusion characteristics of OCPs and summarize reliable markers and morphological changes in OCs during their fusion process, providing guidance for researchers in identifying OCs both in vitro and in vivo. This review focuses on OC formation, characterization, and the roles of OCs beyond resorption in various bone pathophysiological processes. Finally, therapeutic strategies targeting OCs are discussed.
Collapse
Affiliation(s)
- Qianfeng Xiang
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands
| | - Lei Li
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, GA, 3508, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, CT, 3584, The Netherlands
| | - X Frank Walboomers
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands
- Research Institute for Medical Innovation, Radboudumc, Nijmegen, the Netherlands
| | - Jeroen J J P van den Beucken
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands.
| |
Collapse
|
36
|
Qin B, Bao D, Liu Y, Zeng S, Deng K, Liu H, Fu S. Engineered exosomes: a promising strategy for tendon-bone healing. J Adv Res 2024; 64:155-169. [PMID: 37972886 PMCID: PMC11464473 DOI: 10.1016/j.jare.2023.11.011] [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: 09/27/2023] [Revised: 10/24/2023] [Accepted: 11/12/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Due to the spatiotemporal complexity of the composition, structure, and cell population of the tendon-bone interface (TBI), it is difficult to achieve true healing. Recent research is increasingly focusing on engineered exosomes, which are a promising strategy for TBI regeneration. AIM OF REVIEW This review discusses the physiological and pathological characteristics of TBI and the application and limitations of natural exosomes in the field of tendon-bone healing. The definition, loading strategies, and spatiotemporal properties of engineered exosomes were elaborated. We also summarize the application and future research directions of engineered exosomes in the field of tendon-bone healing. KEY SCIENTIFIC CONCEPTS OF REVIEW Engineered exosomes can spatially deliver cargo to targeted sites and temporally realize the sustained release of therapeutic molecules in TBI. This review expounds on the multidifferentiation of engineered exosomes for tendon-bone healing, which effectively improves the biological and biomechanical properties of TBI. Engineered exosomes could be a promising strategy for tendon-bone healing.
Collapse
Affiliation(s)
- Bo Qin
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Dingsu Bao
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646600, China; Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610000, China
| | - Yang Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Shengqiang Zeng
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646600, China; Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610000, China
| | - Kai Deng
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Huan Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646600, China.
| | - Shijie Fu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646600, China.
| |
Collapse
|
37
|
Ye P, Yang Y, Qu Y, Yang W, Tan J, Zhang C, Sun D, Zhang J, Zhao W, Guo S, Song L, Hou T, Zhang Z, Tang Y, Limjunyawong N, Xu J, Dong S, Dou C, Luo F. LL-37 and bisphosphonate co-delivery 3D-scaffold with antimicrobial and antiresorptive activities for bone regeneration. Int J Biol Macromol 2024; 277:134091. [PMID: 39059543 DOI: 10.1016/j.ijbiomac.2024.134091] [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: 02/20/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024]
Abstract
This study introduces a novel 3D scaffold for bone regeneration, composed of silk fibroin, chitosan, nano-hydroxyapatite, LL-37 antimicrobial peptide, and pamidronate. The scaffold addresses a critical need in bone tissue engineering by simultaneously combating bone infections and promoting bone growth. LL-37 was incorporated for its broad-spectrum antimicrobial properties, while pamidronate was included to inhibit bone resorption. The scaffold's porous structure, essential for cell infiltration and nutrient diffusion, was achieved through a freeze-drying process. In vitro assessments using SEM and FTIR confirmed the scaffold's morphology and chemical integrity. Antimicrobial efficacy was tested against pathogens of Staphylococcus aureus (S. aureus) and Pseudomonas aeruginosa (P. aeruginosa). In vivo studies in a murine model of infectious bone defect revealed the scaffold's effectiveness in reducing inflammation and bacterial load, and promoting bone regeneration. RNA sequencing of treated specimens provided insights into the molecular mechanisms underlying these observations, revealing significant gene expression changes related to bone healing and immune response modulation. The results indicate that the scaffold effectively inhibits bacterial growth and supports bone cell functions, making it a promising candidate for treating infectious bone defects. Future studies should focus on optimizing the release of therapeutic agents and evaluating the scaffold's clinical potential.
Collapse
Affiliation(s)
- Peng Ye
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Yusheng Yang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ying Qu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wenxin Yang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiulin Tan
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chengmin Zhang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Dong Sun
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jie Zhang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Weikang Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Shuquan Guo
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Lei Song
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Tianyong Hou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zehua Zhang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yong Tang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Nathachit Limjunyawong
- Center of Research Excellence in Allergy and Immunology, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shiwu Dong
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China.
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Fei Luo
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
38
|
Jiang N, Jiang J, Wang Q, Hao J, Yang R, Tian X, Wang H. Strategic targeting of miR-183 and β-catenin to enhance BMSC stemness in age-related osteoporosis therapy. Sci Rep 2024; 14:21489. [PMID: 39277663 PMCID: PMC11401869 DOI: 10.1038/s41598-024-72474-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024] Open
Abstract
Age-related osteoporosis is a prevalent bone metabolic disorder distinguished by an aberration in the equilibrium between bone formation and resorption. The reduction in the stemness of Bone Marrow Mesenchymal Stem Cells (BMSCs) plays a pivotal role in the onset of this ailment. Comprehending the molecular pathways that govern BMSCs stemness is imperative for delineating the etiology of age-related osteoporosis and devising efficacious treatment modalities. The study utilized single-cell RNA sequencing and miRNA sequencing to investigate the cellular heterogeneity and stemness of BMSCs. Through dual-luciferase reporter assays and functional experiments, the regulatory effect of miR-183 on CTNNB1 (β-catenin) was confirmed. Overexpression and knockdown studies were conducted to explore the impact of miR-183 and β-catenin on stemness-related transcription factors Oct4, Nanog, and Sox2. Cell proliferation assays and osteogenic differentiation experiments were carried out to validate the influence of miR-183 and β-catenin on the stemness properties of BMSCs. Single-cell analysis revealed that β-catenin is highly expressed in both high stemness clusters and terminal differentiation clusters of BMSCs. Overexpression of β-catenin upregulated stemness transcription factors, while its suppression had the opposite effect, indicating a dual regulatory role of β-catenin in maintaining BMSCs stemness and promoting bone differentiation. Furthermore, the confluence of miRNA sequencing analyses and predictions from online databases revealed miR-183 as a potential modulator of BMSCs stemness and a novel upstream regulator of β-catenin. The overexpression of miR-183 effectively diminished the stemness characteristics of BMSCs by suppressing β-catenin, whereas the inhibition of miR-183 augmented stemness. These outcomes align with the observed alterations in the expression levels and functional assessments of transcription factors associated with stemness. This study provides evidence for the essential involvement of β-catenin in preserving the stemness of BMSCs, as well as elucidating the molecular mechanism through which miR-183 selectively targets β-catenin to modulate stemness. These results underscore the potential of miR-183 and β-catenin as molecular targets for augmenting the stemness of BMSCs. This strategy is anticipated to facilitate the restoration of bone microarchitecture and facilitate bone tissue regeneration by addressing potential cellular dysfunctions, thereby presenting novel targets and perspectives for the management of age-related osteoporosis.
Collapse
Affiliation(s)
- Nizhou Jiang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jian Jiang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Quanxiang Wang
- Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Jiayu Hao
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Rui Yang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Xiliang Tian
- The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Hong Wang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China.
| |
Collapse
|
39
|
Xiang T, Yang C, Deng Z, Sun D, Luo F, Chen Y. Krüppel-like factors family in health and disease. MedComm (Beijing) 2024; 5:e723. [PMID: 39263604 PMCID: PMC11387732 DOI: 10.1002/mco2.723] [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: 03/19/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/13/2024] Open
Abstract
Krüppel-like factors (KLFs) are a family of basic transcription factors with three conserved Cys2/His2 zinc finger domains located in their C-terminal regions. It is acknowledged that KLFs exert complicated effects on cell proliferation, differentiation, survival, and responses to stimuli. Dysregulation of KLFs is associated with a range of diseases including cardiovascular disorders, metabolic diseases, autoimmune conditions, cancer, and neurodegenerative diseases. Their multidimensional roles in modulating critical pathways underscore the significance in both physiological and pathological contexts. Recent research also emphasizes their crucial involvement and complex interplay in the skeletal system. Despite the substantial progress in understanding KLFs and their roles in various cellular processes, several research gaps remain. Here, we elucidated the multifaceted capabilities of KLFs on body health and diseases via various compliable signaling pathways. The associations between KLFs and cellular energy metabolism and epigenetic modification during bone reconstruction have also been summarized. This review helps us better understand the coupling effects and their pivotal functions in multiple systems and detailed mechanisms of bone remodeling and develop potential therapeutic strategies for the clinical treatment of pathological diseases by targeting the KLF family.
Collapse
Affiliation(s)
- Tingwen Xiang
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Chuan Yang
- Department of Biomedical Materials Science Third Military Medical University (Army Medical University) Chongqing China
| | - Zihan Deng
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Dong Sun
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Fei Luo
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Yueqi Chen
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
- Department of Orthopedics Chinese PLA 76th Army Corps Hospital Xining China
| |
Collapse
|
40
|
Liu J, Wang B, Chen H, Yu X, Cao X, Zhang H. Osteoclast-derived exosomes influence osteoblast differentiation in osteoporosis progression via the lncRNA AW011738/ miR-24-2-5p/ TREM1 axis. Biomed Pharmacother 2024; 178:117231. [PMID: 39094542 DOI: 10.1016/j.biopha.2024.117231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
AIMS To investigate the molecular mechanism of osteoclast-derived exosomes in osteoporosis. MAIN METHODS RANKL induced osteoclast model was screened for significantly differentially expressed lncRNAs and mRNAs by whole RNA sequencing. Exosomes were characterized using electron microscopy, western blotting and nanosight. Overexpression or knockdown of AW011738 was performed to explore its function. The degree of osteoporosis in an osteoporosis model was assessed by mirco-CT. The osteoclast model, osteoblast differentiation ability and the molecular mechanism of lncRNA AW011738/miR-24-2-5p/TREM1 axis in osteoporosis were assessed by dual luciferase reporter gene assay, Western blotting (WB), immunofluorescence and ALP staining. Bioinformatics was used to predict interactions of key osteoporosis-related genes with miRNAs, transcription factors, and potential drugs after upregulation of AW011738. To predict the protein-protein interaction (PPI) network associated with key genes, GO and KEGG analyses were performed on the key genes. The ssGSVA was used to predict changes in the immune microenvironment. KEY FINDINGS Osteoclast-derived exosomes containing lncRNA AW011738 decreased the osteogenesis-related markers and accelerated bone loss in OVX mice. Osteoclast (si-AW011738)-derived exosomes showed a significant increase in biomarkers of osteoblast differentiation in vitro compared to the si-NC group. As analyzed by mirco-CT, tail vein injected si-AW011738 OVX mice were less osteoporotic than the control group. AW011738 inhibited osteoblast differentiation by regulating TREM1 expression through microRNA. Meanwhile, overexpression of miR-24-2-5p inhibited TREM1 expression to promote osteoblast differentiation. SIGNIFICANCE Osteoclast-derived exosomes containing lncRNA AW011738 inhibit osteogenesis in MC3T3-E1 cells through the lncRNA AW011738/miR-24-2-5p/TREM1 axis and exacerbate osteoporosis in OVX mice.
Collapse
Affiliation(s)
- Jingcheng Liu
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Binyu Wang
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Hongtao Chen
- Department Of Orthopedics, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China.
| | - Xiao Yu
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Xiaojian Cao
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Hongxiu Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
41
|
Zhu P, Tao H, Chen K, Chu M, Wang Q, Yang X, Zhou J, Yang H, Geng D. TRPA1 aggravates osteoclastogenesis and osteoporosis through activating endoplasmic reticulum stress mediated by SRXN1. Cell Death Dis 2024; 15:624. [PMID: 39191723 DOI: 10.1038/s41419-024-07018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Osteoporosis (OP) is a disorder of bone remodeling caused by an imbalance between bone resorption by osteoclasts and bone formation by osteoblasts. Therefore, inhibiting excessive osteoclast activity is one of the promising strategies for treating OP. A major transient receptor potential cation channel, known as transient receptor potential ankyrin 1 (TRPA1), was found to alleviate joint pain and cartilage degeneration in osteoarthritis. However, little research has focused on TRPA1 function in OP. As a result, this study aimed to explore the TRPA1 characteristics and its potential therapeutic function during osteoclastogenesis. The TRPA1 expression gradually increased in the osteoclast differentiation process; however, its suppression with small interfering RNA and an inhibitor (HC030031) significantly controlled the osteoclast count and the expression of osteoclast characteristic genes. Its suppression also inhibited endoplasmic reticulum (ER) stress-related pancreatic ER kinase (PERK) pathways. An ER stress inhibitor (thapsigargin) reversed the down-regulated levels of ER stress and osteoclast differentiation by suppressing TRPA1. Transcriptome sequencing results demonstrated that TRPA1 negatively regulated reactive oxygen species (ROS) and significantly increased the expression of an antioxidant gene, SRXN1. The osteoclast differentiation and the levels of ER stress were enhanced with SRXN1 inhibition. Finally, TRPA1 knockdown targeting macrophages by adeno-associated virus-9 could relieve osteoclast differentiation and osteopenia in ovariectomized mice. In summary, silencing TRPA1 restrained osteoclast differentiation through ROS-mediated down-regulation of ER stress via inhibiting PERK pathways. The study also indicated that TRPA1 might become a prospective treatment target for OP.
Collapse
Affiliation(s)
- Pengfei Zhu
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Huaqiang Tao
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Kai Chen
- Department of Orthopedics, Hai'an People's Hospital, Hai'an, 226600, Jiangsu, China
| | - Miao Chu
- Department of Orthopedics, Yixing People's Hospital, Yixing, 214200, Jiangsu, China
| | - Qiufei Wang
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People's Hospital of Changshu City, Changshu, 215500, Jiangsu, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, 215008, Jiangsu, China.
| | - Jun Zhou
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Huilin Yang
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
42
|
Wan X, Zhang W, Dai L, Chen L. The Role of Extracellular Vesicles in Bone Regeneration and Associated Bone Diseases. Curr Issues Mol Biol 2024; 46:9269-9285. [PMID: 39329900 PMCID: PMC11430372 DOI: 10.3390/cimb46090548] [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: 08/03/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoscale particles with a lipid bilayer membrane structure secreted by various cell types. Nearly all human cells secrete EVs, primarily mediating intercellular communication. In recent years, scientists have discovered that EVs can carry multiple biological cargos, such as DNA, non-coding RNAs (ncRNAs), proteins, cytokines, and lipids, and mediate intercellular signal transduction. Bone is a connective tissue with a nerve supply and high vascularization. The repair process after injury is highly complex, involving interactions among multiple cell types and biological signaling pathways. Bone regeneration consists of a series of coordinated osteoconductive and osteoinductive biological processes. As mediators of intercellular communication, EVs can promote bone regeneration by regulating osteoblast-mediated bone formation, osteoclast-mediated bone resorption, and other pathways. This review summarizes the biogenesis of EVs and the mechanisms by which EV-mediated intercellular communication promotes bone regeneration. Additionally, we focus on the research progress of EVs in various diseases related to bone regeneration. Finally, based on the above research, we explore the clinical applications of engineered EVs in the diagnosis and treatment of bone regeneration-related diseases.
Collapse
Affiliation(s)
- Xinyue Wan
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
| | - Wenjie Zhang
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
| | - Lingyan Dai
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
| | - Liang Chen
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing University School of Medicine, Chongqing 400030, China
| |
Collapse
|
43
|
Guo X, Qin Y, Feng Z, Li H, Yang J, Su K, Mao R, Li J. Investigating the anti-inflammatory effects of icariin: A combined meta-analysis and machine learning study. Heliyon 2024; 10:e35307. [PMID: 39170422 PMCID: PMC11336647 DOI: 10.1016/j.heliyon.2024.e35307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024] Open
Abstract
Objective The objectives of this study were to define the superiority of icariin and its derivatives' anti-inflammatory activities and to create a reference framework for evaluating preclinical evidence. This method combines machine learning and meta-analysis to identify underlying biological pathways. Methods Data came from PubMed, Embase, Web of Science, and the Cochrane Library. SYRCLE was used to evaluate the risk of bias in a subset of research. Meta-analysis and detailed subgroup analyses, categorized by species, genders, disease type, dosage, and treatment duration, were performed using R and STATA 15.0 software to derive nuanced insights. Employing R software (version 4.2.3) and the tidymodels package, the analysis focused on constructing a model and selecting features, with TNF-α as the dependent variable. This approach aims to identify significant predictors of drug efficacy. An in-depth literature facilitated the synthesis of anti-inflammatory mechanisms attributed to icariin and its constituent compounds. Results Following a meticulous search and selection process, 19 studies, involving 370 and 260 animals were included in the meta-analysis and machine-learning assessment, respectively. The findings revealed that icariin and its derivatives markedly reduced inflammation markers, including TNF-α and IL-1β. Additionally, machine-learning outcomes, with TNF-α as the target variable, indicated enhanced anti-inflammatory effects of icariin across respiratory, urological, neurological, and digestive disease types. These effects were more pronounced at doses exceeding 27.52 mg/kg/day and treatment durations beyond 31.22 days. Conclusion Strong anti-inflammatory effects are exhibited by icariiin and its derivatives, which are especially beneficial in the management of digestive, neurological, pulmonary, and urinary conditions. Effective for periods longer than 31.22 days and at dosages more than 27.52 mg/kg/day. Subsequent research will involve more targeted animal experiments and safety assessments to obtain more comprehensive preclinical evidence.
Collapse
Affiliation(s)
- Xiaochuan Guo
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Yanqin Qin
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Zhenzhen Feng
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Haibo Li
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Jingfan Yang
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Kailin Su
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Ruixiao Mao
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| | - Jiansheng Li
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases By Henan and Education Ministry of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan province, China
| |
Collapse
|
44
|
Li M, Zhao Z, Yi J. Biomaterials Designed to Modulate Reactive Oxygen Species for Enhanced Bone Regeneration in Diabetic Conditions. J Funct Biomater 2024; 15:220. [PMID: 39194658 DOI: 10.3390/jfb15080220] [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: 06/02/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Diabetes mellitus, characterized by enduring hyperglycemia, precipitates oxidative stress, engendering a spectrum of complications, notably increased bone vulnerability. The genesis of reactive oxygen species (ROS), a byproduct of oxygen metabolism, instigates oxidative detriment and impairs bone metabolism in diabetic conditions. This review delves into the mechanisms of ROS generation and its impact on bone homeostasis within the context of diabetes. Furthermore, the review summarizes the cutting-edge progress in the development of ROS-neutralizing biomaterials tailored for the amelioration of diabetic osteopathy. These biomaterials are engineered to modulate ROS dynamics, thereby mitigating inflammatory responses and facilitating bone repair. Additionally, the challenges and therapeutic prospects of ROS-targeted biomaterials in clinical application of diabetic bone disease treatment is addressed.
Collapse
Affiliation(s)
- Mingshan Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jianru Yi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
45
|
Zhou S, Wen H, Wang B, Guan S, Fang X. Serum levels of soluble receptor activator for nuclear factor kB ligand play a crucial role in the association of osteoprotegerin with coronary artery disease. Exp Ther Med 2024; 28:325. [PMID: 38979019 PMCID: PMC11229391 DOI: 10.3892/etm.2024.12614] [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: 09/25/2023] [Accepted: 04/08/2024] [Indexed: 07/10/2024] Open
Abstract
Osteoprotegerin (OPG) is a soluble decoy receptor for receptor activator of nuclear factor kB ligand (RANKL), and is implicated in the pathogenesis of atherosclerosis. The aim of the present study was to examine the hypothesis that serum OPG concentrations are increased in patients with stable coronary artery disease (CAD) at different serum levels of soluble RANKL (sRANKL). The study used a case-control design in which consecutively hospitalized individuals were recruited. Fasting blood samples were taken upon admission for serum testing. Participants with previously diagnosed CAD that was asymptomatic or had controlled symptoms constituted the stable CAD group, whereas patients with negative coronary computed tomography angiography results constituted the control non-CAD group. Exclusion criteria included recent acute coronary syndrome, severe heart failure, CAD-complicating autoimmune, blood or thyroid diseases, cancer, elevated temperature with or without infection, severe liver or kidney dysfunction, abnormal calcium metabolism, recent surgery and trauma history. A total of 118 individuals were included in the study. Smoothed plots generated using the recursive method and multivariate models showed that the incidence of stable CAD increased with serum OPG level up to the turning point of 18 pg/ml. This trend was observed at both high [odds ratio (OR), 1.61; 95% confidence interval (CI), 1.04-2.50; P=0.032) and low sRANKL concentrations (OR, 1.52; 95% CI, 1.06-2.17; P=0.022) after adjustment for cardiovascular risk factors. In conclusion, serum OPG levels ≤18 pg/ml are positively associated with stable CAD, regardless of sRANKL levels. In addition, at the same serum OPG level, higher sRANKL levels are associated with a greater incidence of stable CAD compared with lower sRANKL levels. This study identified the relationship between OPG, sRANKL, and stable CAD, and established the reference range for future clinical use.
Collapse
Affiliation(s)
- Shaoqiong Zhou
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hui Wen
- Department of General Practice, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Bin Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Siming Guan
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xin Fang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
46
|
Yang K, Wu Z, Zhang K, Weir MD, Xu HHK, Cheng L, Huang X, Zhou W. Unlocking the potential of stimuli-responsive biomaterials for bone regeneration. Front Pharmacol 2024; 15:1437457. [PMID: 39144636 PMCID: PMC11322102 DOI: 10.3389/fphar.2024.1437457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
Bone defects caused by tumors, osteoarthritis, and osteoporosis attract great attention. Because of outstanding biocompatibility, osteogenesis promotion, and less secondary infection incidence ratio, stimuli-responsive biomaterials are increasingly used to manage this issue. These biomaterials respond to certain stimuli, changing their mechanical properties, shape, or drug release rate accordingly. Thereafter, the activated materials exert instructive or triggering effects on cells and tissues, match the properties of the original bone tissues, establish tight connection with ambient hard tissue, and provide suitable mechanical strength. In this review, basic definitions of different categories of stimuli-responsive biomaterials are presented. Moreover, possible mechanisms, advanced studies, and pros and cons of each classification are discussed and analyzed. This review aims to provide an outlook on the future developments in stimuli-responsive biomaterials.
Collapse
Affiliation(s)
- Ke Yang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhuoshu Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Keke Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Michael D. Weir
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Hockin H. K. Xu
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Wen Zhou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
47
|
Elahmer NR, Wong SK, Mohamed N, Alias E, Chin KY, Muhammad N. Mechanistic Insights and Therapeutic Strategies in Osteoporosis: A Comprehensive Review. Biomedicines 2024; 12:1635. [PMID: 39200100 PMCID: PMC11351389 DOI: 10.3390/biomedicines12081635] [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: 03/13/2024] [Revised: 05/10/2024] [Accepted: 07/16/2024] [Indexed: 09/01/2024] Open
Abstract
Osteoporosis, a metabolic bone disorder characterized by decreased bone mass per unit volume, poses a significant global health burden due to its association with heightened fracture risk and adverse impacts on patients' quality of life. This review synthesizes the current understanding of the pathophysiological mechanisms underlying osteoporosis, with a focus on key regulatory pathways governing osteoblast and osteoclast activities. These pathways include RANK/RANKL/OPG, Wingless-int (Wnt)/β-catenin, and Jagged1/Notch1 signaling, alongside the involvement of parathyroid hormone (PTH) signaling, cytokine networks, and kynurenine in bone remodeling. Pharmacotherapeutic interventions targeting these pathways play a pivotal role in osteoporosis management. Anti-resorptive agents, such as bisphosphonates, estrogen replacement therapy/hormone replacement therapy (ERT/HRT), selective estrogen receptor modulators (SERMs), calcitonin, anti-RANKL antibodies, and cathepsin K inhibitors, aim to mitigate bone resorption. Conversely, anabolic agents, including PTH and anti-sclerostin drugs, stimulate bone formation. In addition to pharmacotherapy, nutritional supplementation with calcium, vitamin D, and vitamin K2 holds promise for osteoporosis prevention. However, despite the availability of therapeutic options, a substantial proportion of osteoporotic patients remain untreated, highlighting the need for improved clinical management strategies. This comprehensive review aims to provide clinicians and researchers with a mechanistic understanding of osteoporosis pathogenesis and the therapeutic mechanisms of existing medications. By elucidating these insights, this review seeks to inform evidence-based decision-making and optimize therapeutic outcomes for patients with osteoporosis.
Collapse
Affiliation(s)
- Nyruz Ramadan Elahmer
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.R.E.); (S.K.W.); (N.M.); (K.-Y.C.)
- Department of Pharmacology, Pharmacy Faculty, Elmergib University, Al Khums 40414, Libya
| | - Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.R.E.); (S.K.W.); (N.M.); (K.-Y.C.)
| | - Norazlina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.R.E.); (S.K.W.); (N.M.); (K.-Y.C.)
| | - Ekram Alias
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.R.E.); (S.K.W.); (N.M.); (K.-Y.C.)
| | - Norliza Muhammad
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.R.E.); (S.K.W.); (N.M.); (K.-Y.C.)
| |
Collapse
|
48
|
Kong X, Tao S, Ji Z, Li J, Li H, Jin J, Zhao Y, Liu J, Zhao F, Chen J, Feng Z, Chen B, Shan Z. FATP2 regulates osteoclastogenesis by increasing lipid metabolism and ROS production. J Bone Miner Res 2024; 39:737-752. [PMID: 38477781 DOI: 10.1093/jbmr/zjae034] [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: 06/27/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024]
Abstract
Lipid metabolism plays a crucial role in maintaining bone homeostasis, particularly in osteoclasts (OCs) formation. Here, we found that the expression level of FATP2, a transporter for long-chain and very-long-chain fatty acids, was significantly upregulated during OC differentiation and in the bone marrow of mice fed a high-fat diet (HFD). Notably, the use of FATP2 siRNA or a specific inhibitor (Lipofermata) resulted in significant inhibition of OC differentiation, while only slightly affecting osteoblasts. In pathological models of bone loss induced by LPS or ovariectomy, in vivo treatment with Lipofermata was able to rescue the loss of bone mass by inhibiting OC differentiation. RNA sequencing revealed that Lipofermata reduced fatty acid β-oxidation and inhibited energy metabolism, while regulating ROS metabolism to decrease ROS production, ultimately inhibiting OC differentiation. Treatment with Lipofermata, either in vivo or in vitro, effectively rescued the overactivation of OCs, indicating that FATP2 regulated OC differentiation by modulating fatty acid uptake and energy metabolism. These findings suggested that targeting FATP2 may represent a promising therapeutic approach for pathological osteoporosis.
Collapse
Affiliation(s)
- Xiangxi Kong
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Siyue Tao
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Zhongyin Ji
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Jie Li
- Department of Orthopaedic Surgery, Ningbo Medical Center Li Huili Hospital, Ningbo, 315100, Zhejiang, China
| | - Hui Li
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Jiayan Jin
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Yihao Zhao
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Junhui Liu
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Jian Chen
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Zhenhua Feng
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Binhui Chen
- Department of Orthopaedic Surgery, Ningbo Medical Center Li Huili Hospital, Ningbo, 315100, Zhejiang, China
| | - Zhi Shan
- Department of Orthopaedic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, 310016, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| |
Collapse
|
49
|
Wang J, Zhao C, Zhao W, Li S. Deficiency of protein phosphatase 5 resists osteoporosis in diabetic mice. Heliyon 2024; 10:e34027. [PMID: 39071657 PMCID: PMC11283048 DOI: 10.1016/j.heliyon.2024.e34027] [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: 02/03/2024] [Revised: 06/23/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Osteoporosis is a common diabetic consequence that negatively affects patients' health and quality of life. Nevertheless, there is mutual interference between clinical drugs intended to regulate blood glucose and bone metabolism. Therefore, it is crucial to look for new treatment targets that effectively control blood glucose and safely protect the bone health of patients with diabetes. In this study, mice given a high-fat diet were shown to be resistant to osteoporosis and diabetes when protein phosphatase 5 (PP5) knockout (KO) mice were used. Serum markers of bone remodeling show that PP5 KO mice are resistant to decreased bone formation and increased bone resorption brought on by diabetes. The absence of PP5 resists the reduction of osteoblast differentiation and the enhancement of osteoclast differentiation in diabetic mice, according to the in vitro osteoblast differentiation of bone mesenchymal stem cells and osteoclast differentiation of bone marrow-derived macrophages. Subsequent investigation revealed that PP5 deficiency increases the expression of the key regulator of osteoblast differentiation, runt-related transcription factor 2, and decreases the activity of the receptor activator of the nuclear factor-κB ligand/osteoprotegerin pathway, a crucial regulatory signaling pathway for osteoclast differentiation. In conclusion, we discovered that PP5 deficiency protects diabetic mice against osteoporosis for the first time.
Collapse
Affiliation(s)
- Jun Wang
- School of Tourism and Cuisine, Yangzhou University, Yangzhou 225127, China
| | - Changyu Zhao
- School of Tourism and Cuisine, Yangzhou University, Yangzhou 225127, China
| | - Wenpeng Zhao
- School of Tourism and Cuisine, Yangzhou University, Yangzhou 225127, China
| | - Songnan Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
50
|
Yang K, Lei S, Qin X, Mai X, Xie W, Yang S, Wang J. Biodegradable polyvinyl alcohol/nano-hydroxyapatite composite membrane enhanced by MXene nanosheets for guided bone regeneration. J Mech Behav Biomed Mater 2024; 155:106540. [PMID: 38615407 DOI: 10.1016/j.jmbbm.2024.106540] [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: 12/26/2023] [Revised: 03/21/2024] [Accepted: 04/07/2024] [Indexed: 04/16/2024]
Abstract
MXene, as a new category of two-dimensional nanomaterials, exhibits a promising prospect in biomedical applications due to its ultrathin structure and morphology, as well as a range of remarkable properties such as biological, chemical, electronic, and optical properties. In this work, different concentrations of MXene (M) were added to polyvinyl alcohol (PVA, P)/nano-hydroxyapatite (n-HA, H) mixed solution, and series of PVA/n-HA/MXene (PHM) composite membranes were obtained by combining sol-gel and freeze-drying processes. Morphology, chemical composition, surface, and mechanical properties of the prepared PHM membranes were characterized by various techniques. Subsequently, the swelling and degradation performances of the composite membranes were tested by swelling and degradation tests. In addition, in vitro studies like cell adhesion, cytotoxicity, proliferation, osteogenic differentiation, and antibacterial properties of MC3T3-E1 were also evaluated. The results showed that the addition of MXene could apparently improve the composite membranes' physicochemical properties, bioactivity, and osteogenic differentiation. Specially, PHM membrane had the best comprehensive properties when the concentration of MXene was set as 2.0% w/v. In a word, the addition of MXene has a positive effect on improving the mechanical properties, osteogenic induction, and antibacterial properties of PH composite membranes, and the prepared PHM composite membranes possess potential applications for guided bone regeneration.
Collapse
Affiliation(s)
- Kefan Yang
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China; State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Siqi Lei
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China; State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Xiaoli Qin
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China; State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Xiaoxue Mai
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China; State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Weibo Xie
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China.
| | - Shengrong Yang
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jinqing Wang
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|