1
|
Wu B, Li M, Yang F, Lu Y, Yi Y, Liu M, Cheng K, Jiang D, Yan B. A stress-driven model for bone density evolution in rats during orthodontic tooth movement. J Mech Behav Biomed Mater 2025; 165:106932. [PMID: 39970840 DOI: 10.1016/j.jmbbm.2025.106932] [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: 12/09/2024] [Revised: 01/26/2025] [Accepted: 02/08/2025] [Indexed: 02/21/2025]
Abstract
Orthodontic bone remodeling simulations offer a scientific foundation for optimizing treatment plans and predicting outcomes in orthodontics. Since alveolar bone exhibits unique regional responses and high sensitivity to tensile and compressive stresses, traditional models often fail to account for these characteristics, limiting their accuracy in predicting the microstructural changes of alveolar bone under external forces. To address this issue, this study proposes a bone remodeling model based on equivalent stress derived from the Mohr strength theory as the mechanical stimulus. The model differentiates tension and compression zones within the alveolar bone and simulates density changes driven by the orthodontic remodeling process: bone formation in tension zone and resorption in compression zone. Orthodontic experiments on rats were conducted to monitor changes in alveolar bone density at 7 and 14 days. Results revealed a density increase of around 3.16% and 9.84% in tension zone and a decrease of approximately 4.86% and 3.61% in compression zone on days 7 and 14, respectively. A comparison between the experimental data and the simulation results of the bone remodeling algorithm demonstrated a consistent trend, validating that the proposed model effectively reflects the dynamic process of bone remodeling. This study provides a new perspective for orthodontic bone remodeling simulations and lays a foundation for further exploration of the mechanisms underlying alveolar bone density changes.
Collapse
Affiliation(s)
- Bin Wu
- College of Mechanical and Electronic Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Mingna Li
- College of Mechanical and Electronic Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Fan Yang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China; State Key Laboratory Cultivation Base of Research. Prevention and Treatment for Oral Diseases, Nanjing, 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Yi Lu
- College of Mechanical and Electronic Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Yang Yi
- College of Mechanical and Electronic Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Mao Liu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China; State Key Laboratory Cultivation Base of Research. Prevention and Treatment for Oral Diseases, Nanjing, 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Ke Cheng
- College of Mechanical Engineering, Southeast University, Nanjing, 211189, China
| | - Di Jiang
- College of Mechanical and Electronic Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| | - Bin Yan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China; State Key Laboratory Cultivation Base of Research. Prevention and Treatment for Oral Diseases, Nanjing, 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China.
| |
Collapse
|
2
|
Geng X, Yao Y, Huang H, Li Q, Wang L, Fan Y. Mechanical and biological characteristics of 3D-printed auxetic structure in bone tissue engineering. J Biomech 2025; 184:112685. [PMID: 40215656 DOI: 10.1016/j.jbiomech.2025.112685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
The auxetic structures are highly effective in bone implants due to their unique deformation characteristics. However, ideal tissue engineering scaffolds must possess suitable mechanical properties and biocompatibility. The biological effects of auxetic structures require further study. In this study, three types of 3D re-entrant honeycomb structures with varying angles of 75°, 90°, and 105° were designed. These structures were fabricated by stereolithography 3D printing technology. Finite element simulations and compression tests were conducted to evaluate their mechanical properties. Scaffolds were inoculated with preosteoblast MC3T3-E1 cells, and cyclic loading was applied to investigate the influence of structural and mechanical stimulation on cell arrangement and proliferation. The results demonstrated that the 75° scaffold exhibited auxetic characteristics in all compression directions and possessed anti-fracture properties. The 75° scaffold also promoted cell proliferation by structural design. Cyclic compression facilitated the nuclear translocation of YAP, further enhancing cell growth. The combination of anti-fracture properties and the promotion of cell proliferation makes auxetic structures highly promising for extensive applications.
Collapse
Affiliation(s)
- Xuezheng Geng
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yan Yao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China; School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Huiwen Huang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qiao Li
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China; School of Engineering Medicine, Beihang University, Beijing 100191, China; State Key Laboratory of Virtual Reality Technology and Systems, Beihang University, Beijing 100191, China.
| | - Lizhen Wang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China; State Key Laboratory of Virtual Reality Technology and Systems, Beihang University, Beijing 100191, China
| | - Yubo Fan
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China; School of Engineering Medicine, Beihang University, Beijing 100191, China; State Key Laboratory of Virtual Reality Technology and Systems, Beihang University, Beijing 100191, China.
| |
Collapse
|
3
|
Li Z, Guo Z, Yang Z, Yang B, Hu Y, Xie X, Zong Z, Chen Z, Zhang K, Zhao P, Li G, Yang X, Bian L. Metabolite-dependent m 6A methylation driven by mechanotransduction-metabolism-epitranscriptomics axis promotes bone development and regeneration. Cell Rep 2025; 44:115611. [PMID: 40272981 DOI: 10.1016/j.celrep.2025.115611] [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: 08/29/2024] [Revised: 03/18/2025] [Accepted: 04/03/2025] [Indexed: 04/26/2025] Open
Abstract
Intramembranous ossification, a major bone development process, begins with the condensation of precursor cells through the timely structural adaption of extracellular matrix (ECM) catering to rapid cellular morphological changes. Inspired by this, we design a highly cell-adaptable hydrogel to recapitulate an ECM-dependent mechanotransduction-metabolism-epitranscriptomics axis in mesenchymal stromal cells (MSCs). This hydrogel significantly enhances the E-cadherin-mediated cell-cell interactions of MSCs and promotes glucose uptake and tricarboxylic acid (TCA) cycle activities. We further show that elevated succinate inhibits fat mass and obesity-associated protein (FTO), a N6-methyladenosine (m6A) demethylase, thereby enhancing methyltransferase-like 3 (METTL3)-driven m6A methylation. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) indicates increased m6A methylation of runt-related transcription 2 (Runx2), a key osteogenic signaling factor, promoting osteogenesis of hydrogel-delivered MSCs and bone regeneration in critical-sized bone defects. Our findings reveal the mechanism underlying the critical impact of adaptable ECM structures on tissue development and provide valuable guidance for the design of ECM-mimetic cell carriers to enhance the therapeutic outcomes of regenerative medicine.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Zhengnan Guo
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Zhengmeng Yang
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Boguang Yang
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Yuan Hu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xian Xie
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Zhixian Zong
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China
| | - Zekun Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, China
| | - Pengchao Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Gang Li
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong 999077, China; Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Xuefeng Yang
- Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei 230601, China.
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
4
|
Mansouri Moghaddam M, Jooybar E, Imani R. Injectable microgel and micro-granular hydrogels for bone tissue engineering. Biofabrication 2025; 17:032001. [PMID: 40228520 DOI: 10.1088/1758-5090/adcc58] [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: 01/17/2025] [Accepted: 04/14/2025] [Indexed: 04/16/2025]
Abstract
Injectable microgels, made from both natural and synthetic materials, are promising platforms for the encapsulation of cells or bioactive agents, such as drugs and growth factors, for delivery to injury sites. They can also serve as effective micro-scaffolds in bone tissue engineering (BTE), offering a supportive environment for cell proliferation or differentiation into osteoblasts. Microgels can be injected in the injury sites individually or in the form of aggregated/jammed ones named micro-granular hydrogels. This review focuses on common materials and fabrication techniques for preparing injectable microgels, as well as their characteristics and applications in BTE. These applications include their use as cell carriers, delivery systems for bioactive molecules, micro-granular hydrogels, bio-inks for bioprinting, three-dimensional microarrays, and the formation of microtissues. Furthermore, we discuss the current and potential future applications of microgels in bone tissue regeneration.
Collapse
Affiliation(s)
- Melika Mansouri Moghaddam
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
5
|
Jin Y, Li J, Fan H, Du J, He Y. Biomechanics and Mechanobiology of Additively Manufactured Porous Load-Bearing Bone Implants. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2409955. [PMID: 40244634 DOI: 10.1002/smll.202409955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/25/2025] [Indexed: 04/18/2025]
Abstract
Given that they can replicate both the biomechanical and mechanobiological functions of natural bone, metal additively manufactured porous load-bearing bone implants present a significant advancement in orthopedic applications. Additive manufacturing (AM) of metals enables precise control over pore geometry, resulting in implants that provide effective mechanical support and minimize stress shielding. In addition to its mechanical benefits, the porous architecture of the implants facilitates essential mechanobiological processes, including the transmission of mechanical signals that regulate cellular processes such as adhesion, proliferation, and differentiation. Before clinical use, the implants should first be engineered to achieve a comparable elastic modulus to native bone, mitigating implant-induced bone resorption while promoting tissue regeneration. It is also noteworthy that the microstructural features of these implants support angiogenesis-a critical process for oxygen and nutrient delivery during bone healing. Despite their potential benefits, challenges remain in balancing mechanical stability for load-bearing applications with biofunctionality for effective integration and controlled degradation. This review comprehensively discusses the biomechanical and mechanobiological factors influencing the design and performance of additively manufactured porous bone implants, highlighting their potential to enhance clinical outcomes in bone repair and regeneration.
Collapse
Affiliation(s)
- Yuan Jin
- Zhejiang-Italy Joint Lab for Smart Materials and Advanced Structures, School of Mechanical Engineering and Mechanics, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Jianhui Li
- Zhejiang-Italy Joint Lab for Smart Materials and Advanced Structures, School of Mechanical Engineering and Mechanics, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Haitao Fan
- Department of Orthopaedics, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315000, China
| | - Jianke Du
- Zhejiang-Italy Joint Lab for Smart Materials and Advanced Structures, School of Mechanical Engineering and Mechanics, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Yong He
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
6
|
Xue B, Xu Z, Li L, Guo K, Mi J, Wu H, Li Y, Xie C, Jin J, Xu J, Jiang C, Gu X, Qin M, Jiang Q, Cao Y, Wang W. Hydrogels with programmed spatiotemporal mechanical cues for stem cell-assisted bone regeneration. Nat Commun 2025; 16:3633. [PMID: 40240370 PMCID: PMC12003706 DOI: 10.1038/s41467-025-59016-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
Hydrogels are extensively utilized in stem cell-based tissue regeneration, providing a supportive environment that facilitates cell survival, differentiation, and integration with surrounding tissues. However, designing hydrogels for regenerating hard tissues like bone presents significant challenges. Here, we introduce macroporous hydrogels with spatiotemporally programmed mechanical properties for stem cell-driven bone regeneration. Using liquid-liquid phase separation and interfacial supramolecular self-assembly of protein fibres, the macroporous structure of hydrogels provide ample space to prevent contact inhibition during proliferation. The rigid protein fibre-coated pore shell provides sustained mechanical cues for guiding osteodifferentiation and protecting against mechanical loads. Temporally, the hydrogel exhibits tunable degradation rates that can synchronize with new tissue deposition to some extent. By integrating localized mechanical heterogeneity, macroporous structures, surface chemistry, and regenerative degradability, we demonstrate the efficacy of these stem cell-encapsulated hydrogels in rabbit and porcine models. This marks a substantial advancement in tailoring the mechanical properties of hydrogels for stem cell-assisted tissue regeneration.
Collapse
Affiliation(s)
- Bin Xue
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| | - Zhengyu Xu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), MOE Key Laboratory of High Performance Polymer Materials and Technology, and State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China
| | - Kaiqiang Guo
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Jing Mi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Haipeng Wu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Yiran Li
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Chunmei Xie
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jing Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Juan Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chunping Jiang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Xiaosong Gu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Meng Qin
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China.
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center (ChemBIC), MOE Key Laboratory of High Performance Polymer Materials and Technology, and State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
7
|
Saldaña L, Vallés G, Vilaboa N, García-Rey E. Microstructural and transcriptomic characterization of trabecular bone in idiopathic osteonecrosis of the femoral head. Sci Rep 2025; 15:11999. [PMID: 40200031 PMCID: PMC11978847 DOI: 10.1038/s41598-025-96726-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
This study aimed to investigate whether idiopathic osteonecrosis of the femoral head (ONFH) is associated with alterations in the microstructure, histological characteristics, and transcriptomic signature in the trabecular region of the femoral head. For this purpose, we obtained trabecular bone explants from the femoral head and the intertrochanteric region of patients with idiopathic ONFH and age- matched patients with primary osteoarthritis (OA). Trabecular bone from the femoral head of ONFH patients showed lower trabecular thickness, bone volume fraction and degree of anisotropy, and a higher percentage of empty lacunae than bone samples from the intertrochanteric region of the same patients and from the femoral head of the OA group. The transcriptome analysis identified a substantial number of genes exclusively regulated in the femoral head of ONFH patients. Among these genes, we found that those highly expressed around the necrotic lesion were involved in cell division and immune response. By contrast, downregulated genes were mainly involved in cell adhesion, angiogenesis and bone formation, such as those encoding collagen type I, bone sialoprotein and several bone morphogenetic proteins. These data add new insights into mechanisms involved in the pathophysiology of idiopathic ONFH.
Collapse
Affiliation(s)
- Laura Saldaña
- Grupo de Fisiopatología Ósea y Biomateriales, Instituto de Investigación del Hospital Universitario La Paz-IdiPAZ, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina- CIBER-BBN, Madrid, Spain.
| | - Gema Vallés
- Grupo de Fisiopatología Ósea y Biomateriales, Instituto de Investigación del Hospital Universitario La Paz-IdiPAZ, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina- CIBER-BBN, Madrid, Spain
| | - Nuria Vilaboa
- Grupo de Fisiopatología Ósea y Biomateriales, Instituto de Investigación del Hospital Universitario La Paz-IdiPAZ, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina- CIBER-BBN, Madrid, Spain
| | - Eduardo García-Rey
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina- CIBER-BBN, Madrid, Spain
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario La Paz-IdiPAZ, Madrid, Spain
| |
Collapse
|
8
|
Cobb C, Wu M, Tangpricha V. Cystic fibrosis-related bone disease: an update on screening, diagnosis, and treatment. Ther Adv Endocrinol Metab 2025; 16:20420188251328210. [PMID: 40183033 PMCID: PMC11967205 DOI: 10.1177/20420188251328210] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Cystic fibrosis-related bone disease (CFBD) is a common endocrinopathy in people living with cystic fibrosis (CF) that is complex and multifactorial in origin. People with CF experience high rates of progressive bone density loss and increased fracture risk. Focus on prevention and treatment of CFBD is of increasing importance in a now aging CF population. This review will discuss current practices in CFBD, gaps in knowledge, and potential future studies with the goal of advancing the clinical care of patients with CFBD.
Collapse
Affiliation(s)
- Crystal Cobb
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Malinda Wu
- Division of Endocrinology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vin Tangpricha
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University School of Medicine, 101 Woodruff Circle, WMRB 1301, Atlanta, GA 30322, USA
- Department of Medicine, Atlanta VA Medical Center, Decatur, GA, USA
| |
Collapse
|
9
|
Zhou Y, Li M, Lin S, Zhu Z, Zhuang Z, Cui S, Chen L, Zhang R, Wang X, Shen B, Chen C, Yang R. Mechanical sensing protein PIEZO1 controls osteoarthritis via glycolysis mediated mesenchymal stem cells-Th17 cells crosstalk. Cell Death Dis 2025; 16:231. [PMID: 40169556 PMCID: PMC11961634 DOI: 10.1038/s41419-025-07577-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025]
Abstract
Aberrant mechanical stimuli can cause tissue attrition and activate mechanosensitive intracellular signaling, impacting the progression of osteoarthritis (OA). However, the precise relationship between mechanical loading and bone metabolism remains unclear. Here, we present evidence that Piezo1 senses the mechanical stimuli to coordinate the crosstalk between mesenchymal stem cells (MSCs) and T helper 17 (Th17) cells, leading to the deterioration of bone and cartilage in osteoarthritis (OA). Mechanical loading impaired the property of MSCs by inhibiting their osteo-chondrogenic differentiation and promoting inflammatory signaling to enhance Th17 cells. Mechanistically, mechanical stimuli activated Piezo1, thereby facilitating Ca2+ influx which upregulated the activity of Hexokinase 2(HK2), the rate-limiting enzyme of glycolysis. The resultant increase in glycolytic activity enhanced communication between MSCs and T cells, thus promoting Th17 cell polarization in a macrophage migration inhibitory factor (MIF) dependent manner. Functionally, Wnt1cre; Piezo1fl/fl mice reduced bone and cartilage erosion in the temporomandibular joint condyle following mechanical loading compared to control groups. Additionally, we observed activated Piezo1 and HK2-mediated glycolysis in patients with temporomandibular joint OA, thereby confirming the clinical relevance of our findings. Overall, our results provide insights into how Piezo1 in MSCs coordinates with mechano-inflammatory signaling to regulate bone metabolism. The schema shows that mechanical sensing protein PIEZO1 in MSCs controls osteoarthritis via glycolysis mediated MSCs and Th17 cells crosstalk in a MIF dependent manner.
Collapse
Affiliation(s)
- Yikun Zhou
- Department of Orthodontics, Peking University School and Hospital of 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 & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
- Department of Orthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mingzhao Li
- Department of Orthodontics, Peking University School and Hospital of 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 & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Shuai Lin
- Department of Orthodontics, Peking University School and Hospital of 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 & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Zilu Zhu
- Department of Orthodontics, Peking University School and Hospital of 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 & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Zimeng Zhuang
- Department of Orthodontics, Peking University School and Hospital of 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 & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Shengjie Cui
- Department of Orthodontics, Peking University School and Hospital of 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 & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Liujing Chen
- Department of Orthodontics, Peking University School and Hospital of 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 & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Ran Zhang
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xuedong Wang
- Department of Orthodontics, Peking University School and Hospital of 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 & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China
| | - Bo Shen
- National Institute of Biological Sciences, Beijing, China, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Ruili Yang
- Department of Orthodontics, Peking University School and Hospital of 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 & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Beijing, Beijing, China.
| |
Collapse
|
10
|
Wang Y, Lee BH, Yang Z, Ho TJ, Ci H, Jackson B, Punshon T, Wang B, Levy J, Ho SP. Chewing-Activated TRPV4/PIEZO1- HIF-1α-Zn Axes in a Rat Periodontal Complex. J Dent Res 2025; 104:398-407. [PMID: 39876056 PMCID: PMC11909774 DOI: 10.1177/00220345241294001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
The upstream mechanobiological pathways that regulate the downstream mineralization rates in periodontal tissues are limitedly understood. Herein, we spatially colocalized and correlated compression and tension strain profiles with the expressions of mechanosensory ion channels (MS-ion) TRPV4 and PIEZO1, biometal zinc, mitochondrial function marker (MFN2), cell senescence indicator (p16), and oxygen status marker hypoxia-inducible factor-1α (HIF-1α) in rats fed hard and soft foods. The observed zinc and related cellular homeostasis in vivo were ascertained by TRPV4 and PIEZO1 agonists and antagonists on human periodontal ligament fibroblasts ex vivo. Four-week-old male Sprague-Dawley rats were fed hard (n = 3) or soft (n = 3) foods for 4 wk (in vivo). Significant changes in alveolar socket and root shapes with decreased periodontal ligament space and increased cementum volume fraction were observed in maxillae on reduced loads (soft food). Reduced loads impaired distally localized compression-stimulated PIEZO1 and mesially localized tension-stimulated TRPV4, decreased mitochondrial function (MFN2), and increased cell senescence in mesial and distal periodontal regions. The switch in HIF-1α from hard food-distal to soft food-mesial indicated a plausible effect of shear-regulated blood and oxygen flows in the periodontal complex. Blunting or activating TRPV4 or PIEZO1 MS-ion channels by channel-specific antagonists or agonists in human periodontal ligament fibroblast cultures (in vitro) indicated a positive correlation between zinc levels and zinc transporters but not with MS-ion channel expressions. The effects of reduced chewing loads in vivo were analogous to TRPV4 and PIEZO1 antagonists in vitro. Study results collectively illustrated that tension-induced TRPV4 and compression-induced PIEZO1 activations are necessary for cell metabolism. An increased hypoxic state with reduced functional loads can be a conducive environment for cementum growth. From a practical standpoint, dose rate-controlled loads can modulate tension and compression-specific MS-ion channel activation, cellular zinc, and HIF-1α transcription. These mechanobiochemical events indicate the plausible catalytic role of biometal zinc in mineralization, periodontal maintenance, and dentoalveolar joint function.
Collapse
Affiliation(s)
- Y Wang
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - B H Lee
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - Z Yang
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - T J Ho
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - H Ci
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- International Research Center for Computational Mechanics, Dalian University of Technology, Dalian, China
- Ningbo Institute of Dalian University of Technology, Ningbo, China
- DUT-BSU Joint Institute, Dalian University of Technology, Dalian, China
| | - B Jackson
- Deparment of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - T Punshon
- Deparment of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - B Wang
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- International Research Center for Computational Mechanics, Dalian University of Technology, Dalian, China
- Ningbo Institute of Dalian University of Technology, Ningbo, China
- DUT-BSU Joint Institute, Dalian University of Technology, Dalian, China
| | - J Levy
- Department of Pathology and Computational Biomedicine, Cedars Sinai, Los Angeles, CA, USA
| | - S P Ho
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
- Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
11
|
Kreller T, Boccaccini AR, Jonitz-Heincke A, Detsch R. Alternating electrical fields to stimulate osteogenic cells and biomimetic calcium phosphate-coated titanium substrates-A combinatorial approach to bone regeneration. BIOMATERIALS ADVANCES 2025; 169:214191. [PMID: 39842166 DOI: 10.1016/j.bioadv.2025.214191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Biophysical stimuli such as alternating electrical fields can mimic endogenous electrical potentials and currents in natural bone. This can help to improve the healing and reconstruction of bone tissue. However, little is known about the combined influence of biomaterials and alternating electric fields on bone cells. Therefore, this study aimed to investigate the impact of both, biomaterials and alternating electric fields, on osteoblast as well as osteoclast differentiation. Initially, either RAW 264.7 or MC3T3-E1 cells were seeded on Ti6Al4V substrates as a load-bearing implant material, modified with biomimetic calcium phosphate (BCP), or uncoated as a reference. The cells were stimulated towards osteoclastic and osteoblastic differentiation via respective growth factors. The effects of BCP substrate modification on cell differentiation were examined after 7 days for RAW 264.7 and after 14 days for MC3T3-E1 cells. In a further series of tests, either RAW 264.7 or MC3T3-E1 cells were seeded on BCP-modified Ti6Al4V substrates, stimulated towards differentiation using growth factors, and further electrically stimulated via alternating electric fields of different voltages and frequencies. In parallel to the first test series RAW 264.7 and MC3T3-E1 cells were stimulated for 7 and 14 days, respectively. Cell morphology was examined via scanning electron microscopy. Cell viabilities were assessed via WST-8 assay. Electrically stimulated MC3T3-E1 cell orientation was evaluated based on fluorescence microscopy images. Marker genes were examined via qPCR. While BCP increased osteoclast-specific gene expression, it had the opposite effect on osteoblast-related genes compared to respective cells seeded on uncoated Ti6Al4V substrates. ES with different parameters showed a broad cellular response due to electrocoupling. While cell viability assessments and gene expression analyses showed clear differences between ES samples and unstimulated controls, only minor cell morphology and orientation differences were observed. Furthermore, there was no clear trend towards a dominant influence of either voltage or frequency as control parameters. Further studies were initiated to investigate the underlying intracellular mechanisms targeted by ES. This work provides an introduction to the targeted control of cellular processes using defined electric fields. The optimization of voltage and frequency could provide therapeutic windows to control specific cellular functions and potentially improve bone regeneration and remodeling processes.
Collapse
Affiliation(s)
- T Kreller
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich Alexander-University Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - A R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich Alexander-University Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - A Jonitz-Heincke
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany
| | - R Detsch
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich Alexander-University Erlangen-Nuremberg, 91058 Erlangen, Germany.
| |
Collapse
|
12
|
IZU Y, ISHIKAWA H, SOETA S. Developmental process and homeostasis of whale long bones lacking medullary cavity using the radius of Antarctic minke whales, Balaenoptera bonaerensis. J Vet Med Sci 2025; 87:336-348. [PMID: 39938892 PMCID: PMC11964860 DOI: 10.1292/jvms.24-0430] [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: 10/25/2024] [Accepted: 02/04/2025] [Indexed: 02/14/2025] Open
Abstract
Whales, Earth's largest mammals and live in water, form long bones without marrow cavities. Body size and mechanical stress impact the bone formation and homeostasis, yet the specific developmental processes remain unclear. Here, we demonstrate the histological changes in whale long bones from fetal to mature stages using the radius of Antarctic minke whales in comparison with domestic cats and cattle. Through intramembranous ossification and remodeling, long bones enlarge their diameters and marrow cavities, respectively. It has been demonstrated that relatively small animals, such as cats, develop the radially arranged "primary osteonal bone," whereas larger animals, including cattle, form "laminar bone" initiated by the formation of circumferentially arranged hypercalcified lines during intramembranous ossification. Here, we demonstrated that whales form laminar bones, transitioning from circumferential in the fetus to radial during postnatal growth, and thereafter cortical bones become compact. After maturation, bone remodeling primarily occurs in the lateral and medial regions of long bones, while the bone layers in the cranial-caudal region never undergo complete resorption. As a result, these layers remain as a wire-netting structure composed of thin bone layers, without forming an open medullary cavity. These data suggest that whales enlarge their long bones through laminar bone formation and form long bones without a marrow cavity by regulating bone resorption areas during the developmental process and in maintaining homeostasis.
Collapse
Affiliation(s)
- Yayoi IZU
- Laboratory of Comparative Cellular Biology, Nippon
Veterinary and Life Science University, Tokyo, Japan
- Department of Laboratory Animal Science, The Faculty of
Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Hajime ISHIKAWA
- Japan Dolphin Center, Kagawa, Japan
- Institute of Cetacean Research, Tokyo, Japan
| | - Satoshi SOETA
- Department of Veterinary Anatomy, Nippon Veterinary and Life
Science University, Tokyo, Japan
| |
Collapse
|
13
|
Pejon TMM, Messias LHD, de Oliveira Nascimento RH, Bertolucci V, Ninomiya AF, Beck WR. Melatonin administration on bone properties of animals under hypoestrogenism: A systematic review. Rev Endocr Metab Disord 2025; 26:279-291. [PMID: 40009141 DOI: 10.1007/s11154-025-09953-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE Hypoestrogenism is associated with loss of bone mass and strength. Melatonin has become a strategy due to its actions on bone tissue. This review summarizes the available data on the effects of chronic melatonin administration on bone tissue in animal models with hypoestrogenism. DATA SOURCES A systematic search of the PubMed, Web of Science, and Scopus, databases up to November 27, 2023, was conducted using specified key terms and Boolean operators (bone AND bones OR bone density OR bone diseases OR osteogenesis OR osteoporosis AND melatonin). STUDY SELECTION only controlled studies in English and with rodents. STUDY DESIGN systematic review. DATA EXTRACTION animals' characteristics (sex and hypoestrogenism confirmation), dose, route, and duration of administration of melatonin, and outcomes from the properties of bone. RESULTS A total of 25 studies were identified after the screening process. In the hypoestrogenic state, melatonin administration improved bone mineral density, bone volume ratio, trabecular number in 19 studies, and maximal load/strength and stiffness test in 7. 4 studies reported improved matrix mineralization in bone marrow mesenchymal stem cells. Melatonin increased the expression of RUNX2 in 9 studies, OCN in 6, and OPG in 4, while decreasing RANKL in 3. In 4 studies the melatonin increased the serum osteocalcin levels. CONCLUSION Chronic administration of melatonin appears to improve the biophysical, biomechanical, molecular, and biochemical properties of bone tissue. These benefits promote an osteogenic effect, making melatonin an efficient strategy to preserve microarchitecture and tissue mass in a state of hypoestrogenism.
Collapse
Affiliation(s)
- Taciane Maria Melges Pejon
- Laboratory of Endocrine Physiology and Physical Exercise, Department of Physiological Sciences, Federal University of São Carlos, Washington Luis, Km 235, São Carlos, São Paulo, 13565-905, Brazil
| | - Leonardo Henrique Dalcheco Messias
- Research Group on Technology Applied to Exercise Physiology-GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, São Paulo, 12916-900, Brazil
| | - Rafael Henrique de Oliveira Nascimento
- Laboratory of Endocrine Physiology and Physical Exercise, Department of Physiological Sciences, Federal University of São Carlos, Washington Luis, Km 235, São Carlos, São Paulo, 13565-905, Brazil
| | - Vanessa Bertolucci
- Research Group on Technology Applied to Exercise Physiology-GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, São Paulo, 12916-900, Brazil
| | - André Felipe Ninomiya
- Research Group on Technology Applied to Exercise Physiology-GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista, São Paulo, 12916-900, Brazil
| | - Wladimir Rafael Beck
- Laboratory of Endocrine Physiology and Physical Exercise, Department of Physiological Sciences, Federal University of São Carlos, Washington Luis, Km 235, São Carlos, São Paulo, 13565-905, Brazil.
| |
Collapse
|
14
|
吴 斌, 胡 可, 杨 帆, 卢 轶, 姜 迪, 易 扬, 严 斌. [Relationship between fluid shear stress in alveolar bone under orthodontic forces and bone remodeling rate]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2025; 43:190-196. [PMID: 40132964 PMCID: PMC11960405 DOI: 10.7518/hxkq.2025.2024288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 02/16/2025] [Indexed: 03/27/2025]
Abstract
OBJECTIVES This study explores the differences in fluid flow within alveolar cancellous bone at various sites under orthodontic forces and elucidates the relationship between fluid shear stress and bone remodeling. These fin-dings lay the groundwork for understanding the biomechanical mechanisms of orthodontic tooth movement. METHODS Stress relaxation tests were performed on human alveolar bone samples to determine material parameters by using the Prony series. An inverse model of alveolar bone was then developed for numerical simulations of fluid-structure interactions to calculate fluid flow within cancellous bone. Meanwhile, a rat model of tooth movement was established to investigate variations in bone remodeling speeds across different regions. RESULTS The microstructural distribution of cancellous alveolar bone was similar in humans and rats. The bone volume fraction and trabecular thickness gradually decreased from root cervical region to root apical region, while the trabecular space gradually increased. Under the influence of orthodontic forces, fluid shear stress within cancellous bone showed spatial variability across different levels, with the highest shear stress occurring at the root apical region, ranging from 0 to 0.936 6 Pa. Additionally, the rat model of tooth movement indicated that bone remodeling occurred more rapidly at the root apical region. CONCLUSIONS Fluid stimulation has a remarkable effect on al-veolar bone remodeling, causing changes in the structure of alveolar bone and ultimately regulating the speed of structu-ral remodeling.
Collapse
|
15
|
Zhang C, Wang S, Meng F, Shu D, Huang H, Zhang Y, Dai S. Comparison of trabecular bone microarchitecture between older males with and without a running habit: A cross-sectional study. J Exerc Sci Fit 2025; 23:83-89. [PMID: 39898366 PMCID: PMC11786814 DOI: 10.1016/j.jesf.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/01/2025] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Objectives Despite its prevalence among seniors, the impact of running on trabecular bone microarchitecture, especially in weight-bearing sites, remains relatively unexplored. This cross-sectional study aimed to investigate the impact of habitual running on bone health, specifically bone mineral density (BMD) and trabecular bone microarchitecture, in male older adults. Methods Twenty-five male recreational runners aged between 50 and 75 years old were recruited in this study (RUN; average running experience 7.5 ± 6.0 years, average monthly running volume 217 ± 120 km), and 25 age matched sedentary older males served as controls (CON). Dual-energy X-ray absorptiometry was used to obtain bone mineral density (BMD) measures at whole-body, bilateral proximal femur as well as lumbar spine for all participants. Magnetic resonance imaging was used to obtain trabecular bone microarchitectural parameters at distal femur and distal tibia for all participants. Results Findings revealed no significant difference in BMD between groups for all measured sites (all p > 0.05; d range 0.013-0.540). However, runners displayed higher bone volume fraction and trabecular thickness at the distal tibia (p = 0.012 and 0.001; 95 % CI of MD [-0.030, -0.004] and [-0.013, -0.004]; d = 0.739 and 1.034, respectively) and higher trabecular thickness at the distal femur (p = 0.002; 95 % CI of MD [-0.010, -0.002]; d = 0.907). Conclusions This study provides critical insights into the relationship between running and bone health in older adults, suggesting regular recreational running may positively influence trabecular bone microarchitecture, potentially enhancing bone strength and reducing fracture risk. These findings pave the way for future research to develop evidence-based exercise recommendations for an aging population.
Collapse
Affiliation(s)
- Chuan Zhang
- School of Physical Education and Sport, Central China Normal University, Wuhan, China
| | | | - Fanjing Meng
- School of Clinical Medicine, Hangzhou Normal University, Hangzhou, China
- The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Dingbo Shu
- Zhejiang University Shaoxing Hospital, Shaoxing, China
| | - Haizhen Huang
- School of Physical Education and Sport, Central China Normal University, Wuhan, China
| | - Yanjing Zhang
- The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Siyu Dai
- School of Clinical Medicine, Hangzhou Normal University, Hangzhou, China
- Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Li Z, Li J, Dai S, Su X, Ren M, He S, Guo Q, Liu F. Effects of Stress on Biological Characteristics and Metabolism of Periodontal Ligament Stem Cells of Deciduous Teeth. Int Dent J 2025; 75:908-920. [PMID: 39370340 PMCID: PMC11976548 DOI: 10.1016/j.identj.2024.09.011] [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: 07/21/2024] [Revised: 08/21/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
INTRODUCTION AND AIMS Periodontal ligament stem cells (PDLSCs) from deciduous teeth (DePDLSCs) can perceive and respond to mechanical signals upon exposure to various environments. The effects of mechanical stress on the biological characteristics and metabolism of DePDLSCs were investigated using in vitro stress loading. METHODS DePDLSCs were subjected to mechanical stresses of different strengths. Cell proliferation, expression of osteogenic/osteoclastic factors, apoptosis, and oxidative stress levels were evaluated using CCK-8 assays, alkaline phosphatase staining, real-time PCR, flow cytometry, and malondialdehyde and superoxide dismutase assays. Liquid chromatography-mass spectrometry was used to perform nontargeted metabolomic detection and analysis. RESULTS Under stresses of 75 and 150 kPa, the expression of osteogenesis-related factors OPG, ALP, and RUNX2 decreased, and the ratio of RANKL/OPG significantly increased. A pressure of 150 kPa induced oxidative stress and caused a significant increase in cell apoptosis. Among the differential metabolites screened from the 150 kPa group, spermine, spermidine, ceramide, phosphatidylethanolamine, lysophosphatidylethanolamine, linoleic acid, and docosatrienoic acid were the most significantly upregulated. The metabolites screened from the 75 kPa group were mainly related to glycerophospholipid and sphingolipid metabolism, oxidative phosphorylation, and mineral absorption, which were common pathways affected in both experimental groups. CONCLUSION A certain degree of mechanical stress can inhibit the proliferative activity and osteogenic differentiation of DePDLSCs, enhance their osteoclast-inducing ability, and cause elevated levels of cell apoptosis and oxidative stress. The metabolic expression profile of DePDLSCs changed significantly under stress. Understanding changes in cellular activity and metabolic reactions may provide an experimental basis for elucidating the role of mechanical stress in root resorption and periodontal tissue remodelling of deciduous teeth. CLINICAL RELEVANCE Mechanical stress may affect periodontal tissue remodeling and root resorption of DePDLSc.
Collapse
Affiliation(s)
- Zhengyang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Jinyi Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Shanshan Dai
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Xuelong Su
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Meiyue Ren
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Shuyang He
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR China
| | - Qingyu Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
| | - Fei Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
17
|
Ma Y, Yang Z, Yu B, Lyu K, Wu J, Chen B, Ma K, Hu Y, Chen D. Biomechanical analysis of axial-radial integrated functional gradient material implants in healthy and osteoporotic bones. Dent Mater J 2025; 44:179-189. [PMID: 39956553 DOI: 10.4012/dmj.2024-222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
People with osteoporosis, common among middle-aged and elderly individuals, often need dental implants. Titanium implants, though generally safe, can cause problems due to their stiffness, especially in osteoporotic bone, leading to fractures. This study aims to identify gradient types that offer improved biological adaptation. This was achieved by comparing the mechanical properties of four new two-dimensional functional gradient materials (FGMs) implants to those of conventional and one-dimensional FGM implants in healthy and osteoporotic bone models. The new FGM implants, with reduced stiffness at the bottom and outer parts, kept strain on cancellous bone within safe limits, reducing fracture risk. Notably, the FGM RA L-H implant maintained strain levels within the optimal range (1,500-3,000 µɛ), promoting bone healing and remodeling. By evaluating the stresses and strains, it was concluded that the FGM RA L-H implant is well adapted to significantly reduce stresses and improve bone recovery in healthy and osteoporotic bones.
Collapse
Affiliation(s)
- Yanzhao Ma
- School of Power and Mechanical Engineering, Wuhan University
| | - Zhexuan Yang
- School of Power and Mechanical Engineering, Wuhan University
| | - Boshen Yu
- School of Power and Mechanical Engineering, Wuhan University
| | - Kun Lyu
- 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
| | - Jian Wu
- Wuhan Second Ship Design and Research Institute
| | - Baohua Chen
- School of Power and Mechanical Engineering, Wuhan University
| | - Kena Ma
- 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
| | - Yiqun Hu
- School of Power and Mechanical Engineering, Wuhan University
| | - Dong Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University
- Department of Implantology, School & Hospital of Stomatology, Wuhan University
| |
Collapse
|
18
|
Wang Q, Chen Y, Ding H, Cai Y, Yuan X, Lv J, Huang J, Huang J, Zhang C, Hong Z, Li H, Huang Y, Lin J, Yuan L, Lin L, Yu S, Zhang C, Lin J, Li W, Chang C, Yang B, Zhang W, Fang X. Optogenetic activation of mechanical nociceptions to enhance implant osseointegration. Nat Commun 2025; 16:3093. [PMID: 40164597 PMCID: PMC11958704 DOI: 10.1038/s41467-025-58336-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Orthopedic implants with high elastic modulus often suffer from poor osseointegration due to stress shielding, a phenomenon that suppresses the expression of intracellular mechanotransduction molecules (IMM) such as focal adhesion kinase (FAK). We find that reduced FAK expression under stress shielding is also mediated by decreased calcitonin gene-related peptide (CGRP) released from Piezo2+ mechanosensitive nerves surrounding the implant. To activate these nerves minimally invasively, we develop a fully implantable, wirelessly rechargeable optogenetic device. In mice engineered to express light-sensitive channels in Piezo2+ neurons, targeted stimulation of the L2-3 dorsal root ganglia (DRG) enhances localized CGRP release near the implant. This CGRP elevation activates the Protein Kinase A (PKA)/FAK signaling pathway in bone marrow mesenchymal stem cells (BMSCs), thereby enhancing osteogenesis and improving osseointegration. Here we show that bioelectronic modulation of mechanosensitive nerves offers a strategy to address implant failure, bridging neuroregulation and bone bioengineering.
Collapse
Affiliation(s)
- Qijin Wang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yang Chen
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Haiqi Ding
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yuanqing Cai
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xuhui Yuan
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianhua Lv
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiagu Huang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiexin Huang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Chaofan Zhang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zihao Hong
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hongyan Li
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ying Huang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiamin Lin
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Lin Yuan
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Lan Lin
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shaolin Yu
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Canhong Zhang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianhua Lin
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Wenbo Li
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Cheng Chang
- Institute of New Materials, Guangdong Academy of Sciences, Guangdong-Hong Kong Joint Laboratory of Modern Surface Engineering Technology, Guangdong Provincial Key Laboratory of Modern Surface Engineering Technology, Guangzhou, Guangdong, PR China
| | - Bin Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Wenming Zhang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Xinyu Fang
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
19
|
Zhang H, Qiao W, Liu Y, Yao X, Zhai Y, Du L. Addressing the challenges of infectious bone defects: a review of recent advances in bifunctional biomaterials. J Nanobiotechnology 2025; 23:257. [PMID: 40158189 PMCID: PMC11954225 DOI: 10.1186/s12951-025-03295-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025] Open
Abstract
Infectious bone defects present a substantial clinical challenge due to the complex interplay between infection control and bone regeneration. These defects often result from trauma, autoimmune diseases, infections, or tumors, requiring a nuanced approach that simultaneously addresses infection and promotes tissue repair. Recent advances in tissue engineering and materials science, particularly in nanomaterials and nano-drug formulations, have led to the development of bifunctional biomaterials with combined osteogenic and antibacterial properties. These materials offer an alternative to traditional bone grafts, minimizing complications such as multiple surgeries, high antibiotic dosages, and lengthy recovery periods. This review examines the repair mechanisms in the infectious microenvironment and highlights various bifunctional biomaterials that foster both anti-infective and osteogenic processes. Emerging design strategies are also discussed to provide a forward-looking perspective on treating infectious bone defects with clinically significant outcomes.
Collapse
Affiliation(s)
- Huaiyuan Zhang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Wenyu Qiao
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yu Liu
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Xizhou Yao
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yonghua Zhai
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Longhai Du
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
20
|
da Silva AF, Lima FJ, Moreira AR, Silva CDN, de Oliveira IB, Callera AF, Porfirio AL, Alves LHV, Tibério IDFLC, Velosa APP, Jorgetti V, Teodoro WR, Lopes FDTQDS. Cigarette Smoke Exposure Leads to Organic and Mineral Bone Component Changes: The Importance of Rho Kinase Function in These Events. Cells 2025; 14:503. [PMID: 40214457 PMCID: PMC11987806 DOI: 10.3390/cells14070503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/19/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025] Open
Abstract
Aberrant Rho-associated kinase function could be associated with increased bone fragility. Since cigarette smoke (CS) exposure promotes the increase in bone fragility due to changes in bone tissue components, this study aimed to investigate how CS exposure could modulate the Rho kinase-associated bone structural changes. Mice were assigned to four groups: control; smoke; control with Rho kinase inhibitor administration; and smoke with a Rho kinase inhibitor. Bone samples were obtained to assess bone histomorphometry analysis, type I collagen composition, and MEPE expression in trabeculae. We observed that CS exposure induced decreased trabecular and osteoid thickness. A concomitant increase in the osteoclastic and erosion surfaces and a decrease in the mineralization surface were observed. Additionally, CS exposure decreased the type I collagen and MEPE expression. Rho kinase inhibitor administration recovered the bone mineralization and the collagen type I deposition. Conclusions: CS exposure increases Rho kinase activity in bone cells, leading to structural changes. The administration of a Rho GTPases inhibitor partially reverses these effects, likely due to the recovery in osteoblast activity.
Collapse
Affiliation(s)
- Alex Ferreira da Silva
- Laboratory of Experimental Therapeutics (LIM-20), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.F.d.S.); (F.J.L.); (A.R.M.); (C.d.N.S.); (A.F.C.); (A.L.P.); (L.H.V.A.); (I.d.F.L.C.T.)
| | - Franciele Jesus Lima
- Laboratory of Experimental Therapeutics (LIM-20), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.F.d.S.); (F.J.L.); (A.R.M.); (C.d.N.S.); (A.F.C.); (A.L.P.); (L.H.V.A.); (I.d.F.L.C.T.)
| | - Alyne Riani Moreira
- Laboratory of Experimental Therapeutics (LIM-20), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.F.d.S.); (F.J.L.); (A.R.M.); (C.d.N.S.); (A.F.C.); (A.L.P.); (L.H.V.A.); (I.d.F.L.C.T.)
| | - Cintia do Nascimento Silva
- Laboratory of Experimental Therapeutics (LIM-20), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.F.d.S.); (F.J.L.); (A.R.M.); (C.d.N.S.); (A.F.C.); (A.L.P.); (L.H.V.A.); (I.d.F.L.C.T.)
| | - Ivone Braga de Oliveira
- Laboratory of Renal Physiopathology (LIM-16), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (I.B.d.O.); (V.J.)
| | - Alexandra Fernandes Callera
- Laboratory of Experimental Therapeutics (LIM-20), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.F.d.S.); (F.J.L.); (A.R.M.); (C.d.N.S.); (A.F.C.); (A.L.P.); (L.H.V.A.); (I.d.F.L.C.T.)
| | - Ana Luiza Porfirio
- Laboratory of Experimental Therapeutics (LIM-20), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.F.d.S.); (F.J.L.); (A.R.M.); (C.d.N.S.); (A.F.C.); (A.L.P.); (L.H.V.A.); (I.d.F.L.C.T.)
| | - Luan Henrique Vasconcelos Alves
- Laboratory of Experimental Therapeutics (LIM-20), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.F.d.S.); (F.J.L.); (A.R.M.); (C.d.N.S.); (A.F.C.); (A.L.P.); (L.H.V.A.); (I.d.F.L.C.T.)
| | - Iolanda de Fátima Lopes Calvo Tibério
- Laboratory of Experimental Therapeutics (LIM-20), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.F.d.S.); (F.J.L.); (A.R.M.); (C.d.N.S.); (A.F.C.); (A.L.P.); (L.H.V.A.); (I.d.F.L.C.T.)
| | - Ana Paula Pereira Velosa
- Rheumatology Division (LIM-17), School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.P.P.V.); (W.R.T.)
| | - Vanda Jorgetti
- Laboratory of Renal Physiopathology (LIM-16), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (I.B.d.O.); (V.J.)
| | - Walcy Rosolia Teodoro
- Rheumatology Division (LIM-17), School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.P.P.V.); (W.R.T.)
| | - Fernanda Degobbi Tibério Quirino Dos Santos Lopes
- Laboratory of Experimental Therapeutics (LIM-20), Department of Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil; (A.F.d.S.); (F.J.L.); (A.R.M.); (C.d.N.S.); (A.F.C.); (A.L.P.); (L.H.V.A.); (I.d.F.L.C.T.)
- Thoracic Surgery Research Laboratory (LIM-61), Division of Thoracic Surgery, Instituto do Coracao do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| |
Collapse
|
21
|
Santoro A, Voto A, Fortino L, Guida R, Laudisio C, Cillo M, D’Ursi AM. Bone Defect Treatment in Regenerative Medicine: Exploring Natural and Synthetic Bone Substitutes. Int J Mol Sci 2025; 26:3085. [PMID: 40243725 PMCID: PMC11988823 DOI: 10.3390/ijms26073085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
In recent years, the management of bone defects in regenerative medicine and orthopedic surgery has been the subject of extensive research efforts. The complexity of fractures and bone loss arising from trauma, degenerative conditions, or congenital disorders necessitates innovative therapeutic strategies to promote effective healing. Although bone tissue exhibits an intrinsic regenerative capacity, extensive fractures and critical-sized defects can severely compromise this process, often requiring bone grafts or substitutes. Tissue engineering approaches within regenerative medicine have introduced novel possibilities for addressing nonunions and challenging bone defects refractory to conventional treatment methods. Key components in this field include stem cells, bioactive growth factors, and biocompatible scaffolds, with a strong focus on advancements in bone substitute materials. Both natural and synthetic substitutes present distinct characteristics and applications. Natural grafts-comprising autologous, allogeneic, and xenogeneic materials-offer biological advantages, while synthetic alternatives, including biodegradable and non-biodegradable biomaterials, provide structural versatility and reduced immunogenicity. This review provides a comprehensive analysis of the diverse bone grafting alternatives utilized in orthopedic surgery, emphasizing recent advancements and persistent challenges. By exploring both natural and synthetic bone substitutes, this work offers an in-depth examination of cutting-edge solutions, fostering further research and innovation in the treatment of complex bone defects.
Collapse
Affiliation(s)
- Angelo Santoro
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
- Scuola di Specializzazione in Farmacia Ospedaliera, Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
| | - Andrea Voto
- Department of Orthopaedics and Traumatology, AORN “San Giuseppe Moscati”, 83100 Avellino, Italy;
| | - Luigi Fortino
- Scuola di Specializzazione in Farmacia Ospedaliera, Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
| | - Raffaella Guida
- Presidio Ospedaliero “Villa Malta” di Sarno, Azienda Sanitaria Locale di Salerno, 84087 Sarno, Italy; (R.G.); (C.L.)
| | - Carolina Laudisio
- Presidio Ospedaliero “Villa Malta” di Sarno, Azienda Sanitaria Locale di Salerno, 84087 Sarno, Italy; (R.G.); (C.L.)
| | - Mariarosaria Cillo
- Dipartimento Farmaceutico, Azienda Sanitaria Locale di Salerno, 84124 Salerno, Italy;
| | - Anna Maria D’Ursi
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
| |
Collapse
|
22
|
周 莹, 张 丹, 吴 立, 王 桂, 母 杰, 崔 成, 石 秀, 董 继, 王 瑜, 许 王, 李 晓. [Epidemiological survey of osteoporosis in Beijing over the past decade: a single-center analysis of dual-energy X-ray absorptiometry scans from 30 599 individuals]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:443-452. [PMID: 40159958 PMCID: PMC11955889 DOI: 10.12122/j.issn.1673-4254.2025.03.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Indexed: 04/02/2025]
Abstract
OBJECTIVES To analyze bone mass distribution and the factors affecting bone mass in a general Chinese Han cohort undergoing physical examinations at our center. METHODS We retrospectively collected the data of bone mineral density (BMD) measurements from 30 599 healthy Han Chinese adults (age≥20 years) who underwent dual-energy X-ray absorptiometry scans at our hospital from July, 2013 to July, 2023. Basic parameters including height, body weight, and gender were recorded, and descriptive statistics and correlation analyses were performed using R software. RESULTS In this cohort, the male individuals had a mean peak BMD of 1.00±0.12 g/cm2 in the lumbar vertebrae, 0.94±0.14 g/cm2 in the femoral neck, and 0.99±0.13 g/cm2 in the total hip, significantly higher than the values in the female individuals [0.99±0.12 g/cm2 in the lumbar vertebrae (P=0.022), 0.79±0.11 g/cm2 in the femoral neck (P<0.001), and 0.88±0.11 g/cm2 in the total hip (P<0.001)]. In the overall cohort, the BMD values of the lumbar spine and femur decreased with age after reaching their peak levels. There was a positive correlation between BMD value and body mass index (BMI) in both male and female individuals. The 2013-2014 period recorded the lowest BMD values in the lumbar, hip, and femoral neck, which tended to increase steadily in the following years (2015-2023). CONCLUSIONS Our data suggest that the BMD values vary among different populations, and future multi-center studies using more accurate BMD detection technology are warranted to capture the variation patterns of BMD with demographic characteristics of specific populations.
Collapse
|
23
|
Estévez M, Batoni E, Cicuéndez M, Bonatti AF, Fernández-Marcelo T, De Maria C, González B, Izquierdo-Barba I, Vozzi G. Fabrication of 3D Biofunctional Magnetic Scaffolds by Combining Fused Deposition Modelling and Inkjet Printing of Superparamagnetic Iron Oxide Nanoparticles. Tissue Eng Regen Med 2025:10.1007/s13770-025-00711-2. [PMID: 40100619 DOI: 10.1007/s13770-025-00711-2] [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: 10/01/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Recently, magnetic composite biomaterials have raised attention in bone tissue engineering as the application of dynamic magnetic fields proved to modulate the proliferation and differentiation of several cell types. METHODS This study presents a novel method to fabricate biofunctional magnetic scaffolds by the deposition of superparamagnetic iron oxide nanoparticles (SPIONs) through thermal Drop-On-Demand inkjet printing on three-dimensional (3D) printed scaffolds. Firstly, 3D scaffolds based on thermoplastic polymeric composed by poly-L-lactic acid/poly-caprolactone/poly(3-hydroxybutyrate-co-3-hydroxyvalerate) were fabricated by Fused Deposition Modelling. Then, in a second step, SPIONs were incorporated onto the surface of the scaffolds by inkjet printing following a designed 2D pattern. RESULTS A complete characterization of the resulting magnetic scaffolds was carried out attending to the surface SPIONs deposits, demonstrating the accuracy and versatility of the production technique, as well as the stability under physiological conditions and the magnetic properties. Biological evaluation with human bone marrow mesenchymal stems cells demonstrated biocompatibility of the scaffolds and increased osteogenic capability under the application of a magnetic field, due to the activation of mechanotransduction processes. CONCLUSION These results show that the developed 3D magnetic biofunctional scaffolds can be a very promising tool for advanced and personalised bone regeneration treatments.
Collapse
Affiliation(s)
- Manuel Estévez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Instituto de Investigación Sanitaria, Universidad Complutense de Madrid, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Elisa Batoni
- Department of Information Engineering, University of Pisa, Via Girolamo Caruso 16, 56122, Pisa, Italy
- Research Center "E. Piaggio", University of Pisa, Via Largo Lucio Lazzarino 1, 56122, Pisa, Italy
| | - Mónica Cicuéndez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Amedeo Franco Bonatti
- Research Center "E. Piaggio", University of Pisa, Via Largo Lucio Lazzarino 1, 56122, Pisa, Italy
| | - Tamara Fernández-Marcelo
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040, Madrid, Spain
| | - Carmelo De Maria
- Department of Information Engineering, University of Pisa, Via Girolamo Caruso 16, 56122, Pisa, Italy.
- Research Center "E. Piaggio", University of Pisa, Via Largo Lucio Lazzarino 1, 56122, Pisa, Italy.
| | - Blanca González
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Instituto de Investigación Sanitaria, Universidad Complutense de Madrid, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029, Madrid, Spain
| | - Isabel Izquierdo-Barba
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Instituto de Investigación Sanitaria, Universidad Complutense de Madrid, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029, Madrid, Spain.
| | - Giovanni Vozzi
- Department of Information Engineering, University of Pisa, Via Girolamo Caruso 16, 56122, Pisa, Italy
- Research Center "E. Piaggio", University of Pisa, Via Largo Lucio Lazzarino 1, 56122, Pisa, Italy
| |
Collapse
|
24
|
Resende-Coelho A, Ali MM, James A, Warren A, Gatrell L, Kadhim I, Fu Q, Xiong J, Onal M, Almeida M. Mitochondrial oxidative stress or decreased autophagy in osteoblast lineage cells is not sufficient to mimic the deleterious effects of aging on bone mechanoresponsiveness. Aging (Albany NY) 2025; 17:610-629. [PMID: 40105873 PMCID: PMC11984430 DOI: 10.18632/aging.206213] [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: 06/07/2024] [Accepted: 01/23/2025] [Indexed: 03/20/2025]
Abstract
Exercise-induced mechanical load stimulates bone cells, including osteocytes, to promote bone formation. The bone response to loading is less effective with aging, but the cellular and molecular mechanisms responsible for the impaired mechanoresponsiveness remain unclear. Excessive mitochondrial reactive oxygen species (mtROS) and deficient autophagy are common aging mechanisms implicated in decreased bone formation in old mice. Here, we confirmed that the osteogenic effects of tibia compressive loading are lower in old versus young female mice. We also examined whether an increase in mtROS or decreased autophagy in osteoblast-lineage cells of adult female mice could mimic the deleterious effects of aging. To this end, we loaded mice lacking the antioxidant enzyme superoxide dismutase 2 (Sod2) or autophagy-related 7 (Atg7) in cells targeted by Osterix1 (Osx1)-Cre. Osteocytes in Atg7ΔOsx1 exhibited altered morphology and decreased osteocyte dendrite projections. Two weeks of loading increased cortical bone mass and bone formation rate at both periosteal and endosteal surfaces of Osx1-Cre control mice. Nonetheless, in both Atg7ΔOsx1 and Sod2ΔOsx1 mice the response to loading was identical to that observed in control mice, indicating that compromised Atg7-dependent autophagy or excessive mtROS are not sufficient to impair the bone response to tibial compressive loading. Thus, alternative mechanisms of aging might be responsible for the decreased response of the aged skeleton to mechanical stimuli. These findings also suggest that an intact osteocyte dendrite network is not required for the osteogenic response in this model of bone loading.
Collapse
Affiliation(s)
- Ana Resende-Coelho
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Md Mohsin Ali
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alicen James
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Aaron Warren
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Landon Gatrell
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ilham Kadhim
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Qiang Fu
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melda Onal
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
25
|
Yu J, Ji L, Liu Y, Wang X, Wang J, Liu C. Bone-brain interaction: mechanisms and potential intervention strategies of biomaterials. Bone Res 2025; 13:38. [PMID: 40097409 PMCID: PMC11914511 DOI: 10.1038/s41413-025-00404-5] [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: 09/21/2024] [Revised: 12/02/2024] [Accepted: 12/31/2024] [Indexed: 03/19/2025] Open
Abstract
Following the discovery of bone as an endocrine organ with systemic influence, bone-brain interaction has emerged as a research hotspot, unveiling complex bidirectional communication between bone and brain. Studies indicate that bone and brain can influence each other's homeostasis via multiple pathways, yet there is a dearth of systematic reviews in this area. This review comprehensively examines interactions across three key areas: the influence of bone-derived factors on brain function, the effects of brain-related diseases or injuries (BRDI) on bone health, and the concept of skeletal interoception. Additionally, the review discusses innovative approaches in biomaterial design inspired by bone-brain interaction mechanisms, aiming to facilitate bone-brain interactions through materiobiological effects to aid in the treatment of neurodegenerative and bone-related diseases. Notably, the integration of artificial intelligence (AI) in biomaterial design is highlighted, showcasing AI's role in expediting the formulation of effective and targeted treatment strategies. In conclusion, this review offers vital insights into the mechanisms of bone-brain interaction and suggests advanced approaches to harness these interactions in clinical practice. These insights offer promising avenues for preventing and treating complex diseases impacting the skeleton and brain, underscoring the potential of interdisciplinary approaches in enhancing human health.
Collapse
Affiliation(s)
- Jiaze Yu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Luli Ji
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yongxian Liu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xiaogang Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China.
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China.
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China.
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China.
| | - Changsheng Liu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China.
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China.
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China.
| |
Collapse
|
26
|
Qiu D, Yan B, Xue H, Xu Z, Tan G, Liu Y. Perspectives of exosomal ncRNAs in the treatment of bone metabolic diseases: Focusing on osteoporosis, osteoarthritis, and rheumatoid arthritis. Exp Cell Res 2025; 446:114457. [PMID: 39986599 DOI: 10.1016/j.yexcr.2025.114457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/13/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Bone metabolic disorders, constituting a group of prevalent and grave conditions, currently have a scarcity of therapeutic alternatives. Over the recent past, exosomes have been at the forefront of research interest, owing to their nanoparticulate nature and potential for therapeutic intervention. ncRNAs are a class of heterogeneous transcripts that they lack protein-encoding capacity, yet they can modulate the expression of other genes through multiple mechanisms. Mounting evidence underscores the intricate role of exosomes as ncRNAs couriers implicated in the pathogenesis of bone metabolic disorders. In this review, we endeavor to elucidate recent insights into the roles of three ncRNAs - miRNAs, lncRNAs, and circRNAs - in bone metabolic ailments such as osteoporosis, osteoarthritis, and rheumatoid arthritis. Additionally, we examine the viability of exosomal ncRNAs as innovative, cell-free modalities in the diagnosis and therapeutic management of bone metabolic disorders. We aim to uncover the critical function of exosomal ncRNAs within the context of bone metabolic diseases.
Collapse
Affiliation(s)
- Daodi Qiu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Binghan Yan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Haipeng Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhanwang Xu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Guoqing Tan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yajuan Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250300, China.
| |
Collapse
|
27
|
Sun Q, Li CH, Liu QS, Zhang YB, Hu BS, Feng Q, Lang Y. Research status of biomaterials based on physical signals for bone injury repair. Regen Ther 2025; 28:544-557. [PMID: 40027992 PMCID: PMC11872413 DOI: 10.1016/j.reth.2025.01.025] [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: 10/08/2024] [Revised: 01/02/2025] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Bone defects repair continues to be a significant challenge facing the world. Biological scaffolds, bioactive molecules, and cells are the three major elements of bone tissue engineering, which have been widely used in bone regeneration therapy, especially with the rise of bioactive molecules in recent years. According to their physical properties, they can be divided into force, magnetic field (MF), electric field (EF), ultrasonic wave, light, heat, etc. However, the transmission of bioactive molecules has obvious shortcomings that hinder the development of the tissue-rearing process. This paper reviews the mechanism of physical signal induction in bone tissue engineering in recent years. It summarizes the application strategies of physical signal in bone tissue engineering, including biomaterial designs, physical signal loading strategies and related pathways. Finally, the ongoing challenges and prospects for the future are discussed.
Collapse
Affiliation(s)
- Qi Sun
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Chao-Hua Li
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Qi-Shun Liu
- Department of Orthopedics, Zhejiang Medical & Health Group Hangzhou Hospital, Hangzhou, 310015, China
| | - Yuan-Bin Zhang
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Bai-Song Hu
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Qi Feng
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Yong Lang
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| |
Collapse
|
28
|
Mukhopadhaya J, Bhadani JS. Fixation Failure in Osteoporotic Bone: A Review of Complications and Outcomes. Indian J Orthop 2025; 59:389-404. [PMID: 40201917 PMCID: PMC11973034 DOI: 10.1007/s43465-024-01316-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/11/2024] [Indexed: 04/10/2025]
Abstract
Background Osteoporotic bone poses significant challenges for fixation of fractures due to its compromised bone quality. This issue impacts patient outcomes and, necessitate proper understanding of the biomechanical limitations and the adequacy of current fixation devices. Objective This article aims to address the gaps in literature by examining both the biomechanical and biological factors that contribute to fixation failure in osteoporotic bone, and by analyzing the limitations of current management strategies, with the aim of identifying effective interventions for this vulnerable patient group. What is Already Known Literature acknowledges that osteoporotic bones have reduced bone density and compromised structural integrity, making fixation devices less effective. Fixation failure frequently occurs in these patients due to diminished bone strength and insufficient fixation support, which collectively hinder optimal stabilization and healing. Gap in Literature Despite recognition of the high failure rates associated with osteoporotic bone fixation, there is limited literature detailing a comprehensive approach that integrates biomechanical, biological, and technological advancements to improve fixation outcomes. This article reviews current diagnostic techniques and explores potential innovations in materials and regenerative strategies aimed at enhancing fixation success. Which also guide us about need for future research to focus on developing and validating multifaceted approaches that combine advanced fixation materials and bone regeneration technologies to mitigate failure risks and improve patient outcomes. Conclusion With increasing life expectancy, the incidence of osteoporosis and hence osteoporotic fractures steadily increasing there are multiple fractures which are responsible for this, however as orthopedic surgeon we are required to deal with these fractures in increasing numbers so we need to develop a comprehensive approach to prevention of these fractures' adequate treatment and also the prevention of refractures which are far too common.
Collapse
Affiliation(s)
- John Mukhopadhaya
- Department of Orthopaedics, Paras HMRI Hospital, Patna, Bihar 800014 India
| | | |
Collapse
|
29
|
Zhang Z, Xiang J, Xiao C, Zhu R, Zheng Y, Yi Y, Wang J, Xiong X. Condylar osseous changes following conservative therapies: A cone-beam computed tomography longitudinal study on adult patients with degenerative temporomandibular joint disease. J Craniomaxillofac Surg 2025; 53:262-270. [PMID: 39672699 DOI: 10.1016/j.jcms.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024] Open
Abstract
This retrospective study aimed to comprehensively investigate the impact of non-surgical treatments on condylar osseous changes in adult patients with degenerative joint disease (DJD). Radiographic and clinical data were collected for analysis. Cone-beam computed tomography (CBCT) was used to diagnose DJD, including flattening, erosion, osteophytes, sclerosis and cysts. Condylar osseous changes were divided into three classifications: progression, stability and remission. Kaplan-Meier analyses were performed to evaluate progression-free probability. Hazard ratios (HRs) of overall and specific DJD progression were calculated with multivariate Cox analysis. Hyaluronic acid (HA) injection significantly reduced the progression-free probability (P = 0.0312). HRs of progression after stabilization splint (SS) treatment within one year, glucosamine treatment within six months and HA injection was 3.41 (95% CI: 1.70-6.84; P = 0.0005), 0.35 (95% CI: 0.14-0.86; P = 0.0226), and 1.84 (95% CI: 1.06-3.20; P = 0.0313), respectively. HRs of progression of flattening, erosion, osteophyte, and sclerosis adjusted for gender and age after HA injection were 2.77 (95% CI: 1.32-5.81; P = 0.0071), 2.12 (95% CI: 1.09-4.10; P = 0.0264), 3.38 (95% CI: 1.08-10.54; P = 0.0361) and 2.78 (95% CI: 1.02-7.52; P = 0.0447) respectively. HA injection and SS treatment were possible risk factors for TMJ deterioration, while glucosamine treatment was possible protective factor against TMJ deterioration.
Collapse
Affiliation(s)
- Zihan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jie Xiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chuqiao Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunhao Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yating Yi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Xin Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
30
|
Jain S, Vohora D. Primary components of MCT ketogenic diet are detrimental to bone loss associated with accelerated aging and age-related neurotoxicity in mice. Bone 2025; 192:117383. [PMID: 39732448 DOI: 10.1016/j.bone.2024.117383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Medium chained triglycerides (MCT) ketogenic diet is being extensively investigated for its neuroprotective effects against adverse effects associated with aging and neurodegenerative disorders. Aging is a common risk factor for the development of both osteoporosis and neurological disorders. Hence, suppression of aging and age-related neurodegeneration might contribute to delaying skeletal aging. The present study was designed to investigate the effects of the primary components of the MCT diet, against bone resorption associated with D-gal-induced accelerated aging and D-gal /AlCl3-induced age-related toxicity. We report bone loss in accelerated aged mice and age-related neurotoxic mice through declined Sirtuin1 (SIRT1) expression, depleted bone turnover markers, (P1NP and β-CTX-1), low bone mineral density (BMD), and deterioration of trabecular bone microarchitecture in both the distal femur and proximal tibia bones. Administration of MCT dietary components decanoic acid and octanoic acid, led to a decrease in body weight and only octanoic acid increased serum levels of ketone body, β-hydroxybutyrate (β-HB), but both of them failed to reverse the diminishing effects on bone health associated with aging and age-related neurotoxicity. Surprisingly, decanoic acid, octanoic acid, and their combination also exhibited negative effects on trabecular bone microarchitecture and BMD in the distal femur and proximal tibia bones of healthy mice. The findings from this study provide supporting evidence on the deterioration of bone health associated with aging and age-related neurotoxicity, and the bone resorption potential of MCT dietary supplements that are being prescribed in healthy older populations and elderly persons diagnosed with neurological disorders.
Collapse
Affiliation(s)
- Shreshta Jain
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Divya Vohora
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
31
|
Varela D, Varela T, Conceição N, Cancela ML. Epigenetic Regulation of ZNF687 by miR-142a-3p and DNA Methylation During Osteoblast Differentiation and Mice Bone Development and Aging. Int J Mol Sci 2025; 26:2069. [PMID: 40076693 PMCID: PMC11899743 DOI: 10.3390/ijms26052069] [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: 01/14/2025] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Zinc finger protein 687 (ZNF687), a transcription factor implicated in osteoblast/osteoclast differentiation and linked to Paget's disease of bone, has unclear mechanisms in bone metabolism. Epigenetic disruptions can affect bone cell activity and contribute to bone-related diseases. This work aimed to elucidate the regulatory role of epigenetics in modulating Zfp687 expression throughout osteoblast differentiation and bone growth/aging in mice. Differentiation of the mouse-derived osteoblast precursor cell line (MC3T3-E1) showed increased expression of osteogenic markers and decreased Zfp687 expression. In the hindlimb bones of C57BL/6J mice, the expression of most bone-forming genes decreased from youth to adulthood, while Zfp687 and Runx2 expression was maintained, being only significantly reduced in old mice in comparison to young mice. Bisulfite sequencing revealed hypomethylation of the Zfp687 promoter during MC3T3-E1 differentiation and bone growth/aging. Bioinformatics predicted miR-142a-3p, miR-122b-5p, and miR-124-3p binding sites in Zfp687 3'UTR, and RT-qPCR analysis showed higher expression of these miRNAs in mature osteoblasts. Transfection of a miR-142-3p mimic reduced luciferase activity in the wildtype Zfp687 3'UTR but not the mutant 3'UTR and downregulated the Zfp687 gene and protein levels. In conclusion, miR-142a-3p directly targets the Zfp687 3'UTR, promoting its downregulation during osteoblastogenesis. Furthermore, DNA methylation does not appear to regulate Zfp687 during osteoblast differentiation or bone development in mice.
Collapse
Affiliation(s)
- Débora Varela
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal; (D.V.); (T.V.)
- Faculty of Medicine and Biomedical Sciences, University of Algarve, 8005-139 Faro, Portugal
| | - Tatiana Varela
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal; (D.V.); (T.V.)
- Faculty of Medicine and Biomedical Sciences, University of Algarve, 8005-139 Faro, Portugal
| | - Natércia Conceição
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal; (D.V.); (T.V.)
- Faculty of Medicine and Biomedical Sciences, University of Algarve, 8005-139 Faro, Portugal
- Algarve Biomedical Center, University of Algarve, 8005-139 Faro, Portugal
| | - M. Leonor Cancela
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal; (D.V.); (T.V.)
- Faculty of Medicine and Biomedical Sciences, University of Algarve, 8005-139 Faro, Portugal
- Algarve Biomedical Center, University of Algarve, 8005-139 Faro, Portugal
| |
Collapse
|
32
|
Ye T, Yan J, Kan T, Xie G, Zhang Z, Yin W, Zhao B, Yu Z, Chu L. Articular cartilage degeneration and aberrant osteocyte perilacunar/canalicular remodeling in subchondral bone of patients with developmental dysplasia of the hip. BMC Musculoskelet Disord 2025; 26:165. [PMID: 39966795 PMCID: PMC11837434 DOI: 10.1186/s12891-025-08419-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Developmental dysplasia of the hip (DDH) is a congenital musculoskeletal disease that impairs the hip joint and exacerbates hip osteoarthritis. This study aims to investigate the alterations of osteocytic characteristics including apoptosis, lacuna-canalicular network, and perilacunar/canalicular remodeling (PLR) activity in subchondral bone from DDH patients, and potential relationship of these alterations between the cartilage degeneration and DDH progression. METHODS The femoral head specimens were acquired from 16 patients with hip fractures who received total hip arthroplasty operation, 24 patients with primary hip OA and 25 patients with DDH. The femoral head were scanned by a micro-computed tomography and the volume of interest was used for a micro-finite element analysis. Histological and immunohistochemical staining was used to observe chondrocytes in cartilage and osteocytes in subchondral bone. Terminal deoxynucleotidyl transferase dUTP nick end labeling staining was used to investigate the apoptotic osteocytes in subchondral bone. Ploton silver staining was used to visualize lacunocanalicular network and picrosirius red staining was to visualize collagen fiber orientation in subchondral bone. RESULTS The DDH group showed the highest apoptosis rate of osteocytes and increased PLR activity among the three groups. The micro-finite-element analysis revealed that DDH group had deteriorative microstructural and biomechanical properties of subchondral bone. The histological and immunohistochemical analyses showed that the cartilage degeneration in DDH group was the most severe. Linear regression analysis revealed a significant correlation between osteocytic activity in subchondral bone and cartilage degeneration in DDH. CONCLUSIONS Our findings indicate that the abnormal osteocyte activity in subchondral bone might contribute to the deterioration of subchondral bone structure, which accelerates cartilage degeneration and DDH progression. Targeting subchondral bone remodeling could offer a promising therapeutic strategy for DDH.
Collapse
Affiliation(s)
- Teng Ye
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiren Yan
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyou Kan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Guoming Xie
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhichang Zhang
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjing Yin
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bizeng Zhao
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Linyang Chu
- Department of Sports Medicine, National Center for Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
33
|
Kontogianni GI, Loukelis K, Bonatti AF, Batoni E, De Maria C, Vozzi G, Naseem R, Dalgarno K, Shin H, Vitale-Brovarone C, Chatzinikolaidou M. A Mechanically Stimulated Co-culture in 3-Dimensional Composite Scaffolds Promotes Osteogenic and Anti-osteoclastogenic Activity and M2 Macrophage Polarization. Biomater Res 2025; 29:0135. [PMID: 39911306 PMCID: PMC11794764 DOI: 10.34133/bmr.0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 02/07/2025] Open
Abstract
Bone is subjected to a plethora of mechanical stresses, which have been found to directly influence the equilibrium between bone resorption and formation. Taking this into account, we present herein a novel biomimicking 3-dimensional model that applies cyclic uniaxial compression onto cells co-cultured on 3-dimensionally printed scaffolds consisting of poly L-lactic acid/poly(ε-caprolactone)/poly(3-hydroxybutyrate-co-3-hydroxyvalerate)/Sr-nanohydroxyapatite. The aim is to investigate how compression can modulate the balance between osteogenesis and osteoclastogenesis in co-culture, as well as the polarization of macrophages. One of the key aspects of the current study is the unprecedented development of a growth-factor-free co-culture, sustainable solely by the cross talk between human bone marrow mesenchymal stem cells and human peripheral blood mononuclear cells for their survival and osteogenic/osteoclastogenic differentiation capacity, respectively. Real-time polymerase chain reaction gene expression analysis of the mechanically stimulated constructs revealed up-regulation of the osteogenesis-related markers osteocalcin, osteoprotegerin, and runt-related transcription factor 2, with concurrent down-regulation of the osteoclastogenic markers dendritic-cell-specific transmembrane protein, nuclear factor of activated T cells 1, and tartrate acid phosphatase. The secretion of the receptor activator of nuclear factor kappa-Β ligand and macrophage colony-stimulating factor, as determined from enzyme-linked immunosorbent assay, was also found to depict lower levels compared to static conditions. Finally, macrophage polarization was examined via confocal imaging of tumor necrosis factor-α and interleukin-10 secretion levels, as well as through nitric oxide synthase and arginase 1 markers' gene expression, with the results indicating stronger commitment toward the M2 phenotype after mechanical stimulation.
Collapse
Affiliation(s)
| | - Konstantinos Loukelis
- Department of Materials Science and Engineering,
University of Crete, 70013 Heraklion, Greece
| | - Amedeo Franco Bonatti
- Research Center E. Piaggio, Department of Information Engineering,
University of Pisa, 56126 Pisa, Italy
| | - Elisa Batoni
- Research Center E. Piaggio, Department of Information Engineering,
University of Pisa, 56126 Pisa, Italy
| | - Carmelo De Maria
- Research Center E. Piaggio, Department of Information Engineering,
University of Pisa, 56126 Pisa, Italy
| | - Giovanni Vozzi
- Research Center E. Piaggio, Department of Information Engineering,
University of Pisa, 56126 Pisa, Italy
| | - Raasti Naseem
- School of Engineering,
Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Kenneth Dalgarno
- School of Engineering,
Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Heungsoo Shin
- Department of Bioengineering,
Hanyang University, Seoul 04763, Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader,
Hanyang University, Seoul 04763, Korea
| | | | - Maria Chatzinikolaidou
- Department of Materials Science and Engineering,
University of Crete, 70013 Heraklion, Greece
- Foundation for Research and Technology Hellas (FORTH)-IESL, 70013 Heraklion, Greece
| |
Collapse
|
34
|
Dong Y, Hu Y, Hu X, Wang L, Shen X, Tian H, Li M, Luo Z, Cai C. Synthetic nanointerfacial bioengineering of Ti implants: on-demand regulation of implant-bone interactions for enhancing osseointegration. MATERIALS HORIZONS 2025; 12:694-718. [PMID: 39480512 DOI: 10.1039/d4mh01237b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Titanium and its alloys are the most commonly used biometals for developing orthopedic implants to treat various forms of bone fractures and defects, but their clinical performance is still challenged by the unfavorable mechanical and biological interactions at the implant-tissue interface, which substantially impede bone healing at the defects and reduce the quality of regenerated bones. Moreover, the impaired osteogenesis capacity of patients under certain pathological conditions such as diabetes and osteoporosis may further impair the osseointegration of Ti-based implants and increase the risk of treatment failure. To address these issues, various modification strategies have been developed to regulate the implant-bone interactions for improving bone growth and remodeling in situ. In this review, we provide a comprehensive analysis on the state-of-the-art synthetic nanointerfacial bioengineering strategies for designing Ti-based biofunctional orthopedic implants, with special emphasis on the contributions to (1) promotion of new bone formation and binding at the implant-bone interface, (2) bacterial elimination for preventing peri-implant infection and (3) overcoming osseointegration resistance induced by degenerative bone diseases. Furthermore, a perspective is included to discuss the challenges and potential opportunities for the interfacial engineering of Ti implants in a translational perspective. Overall, it is envisioned that the insights in this review may guide future research in the area of biometallic orthopedic implants for improving bone repair with enhanced efficacy and safety.
Collapse
Affiliation(s)
- Yilong Dong
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325016, China.
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Xinqiang Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Lingshuang Wang
- School of Life Sciences, Chongqing University, Chongqing, 400044, China.
| | - Xinkun Shen
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325016, China.
| | - Hao Tian
- Kairui Stomatological Hospital, Chengdu 610211, China
| | - Menghuan Li
- School of Life Sciences, Chongqing University, Chongqing, 400044, China.
| | - Zhong Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
- School of Life Sciences, Chongqing University, Chongqing, 400044, China.
| | - Chunyuan Cai
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325016, China.
| |
Collapse
|
35
|
Ma WWS, Yang H, Zhao Y, Li X, Ding J, Qu S, Liu Q, Hu Z, Li R, Tao Q, Mo H, Zhai W, Song X. Multi-Physical Lattice Metamaterials Enabled by Additive Manufacturing: Design Principles, Interaction Mechanisms, and Multifunctional Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405835. [PMID: 39834122 PMCID: PMC11848643 DOI: 10.1002/advs.202405835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/21/2024] [Indexed: 01/22/2025]
Abstract
Lattice metamaterials emerge as advanced architected materials with superior physical properties and significant potential for lightweight applications. Recent developments in additive manufacturing (AM) techniques facilitate the manufacturing of lattice metamaterials with intricate microarchitectures and promote their applications in multi-physical scenarios. Previous reviews on lattice metamaterials have largely focused on a specific/single physical field, with limited discussion on their multi-physical properties, interaction mechanisms, and multifunctional applications. Accordingly, this article critically reviews the design principles, structure-mechanism-property relationships, interaction mechanisms, and multifunctional applications of multi-physical lattice metamaterials enabled by AM techniques. First, lattice metamaterials are categorized into homogeneous lattices, inhomogeneous lattices, and other forms, whose design principles and AM processes are critically discussed, including the benefits and drawbacks of different AM techniques for fabricating different types of lattices. Subsequently, the structure-mechanism-property relationships and interaction mechanisms of lattice metamaterials in a range of physical fields, including mechanical, acoustic, electromagnetic/optical, and thermal disciplines, are summarized to reveal critical design principles. Moreover, the multifunctional applications of lattice metamaterials, such as sound absorbers, insulators, and manipulators, sensors, actuators, and soft robots, thermal management, invisible cloaks, and biomedical implants, are enumerated. These design principles and structure-mechanism-property relationships provide effective design guidelines for lattice metamaterials in multifunctional applications.
Collapse
Affiliation(s)
- Winston Wai Shing Ma
- Department of Mechanical and Automation EngineeringChinese University of Hong KongSha TinHong Kong999077China
| | - Hang Yang
- Department of Mechanical EngineeringNational University of SingaporeSingapore117575Singapore
| | - Yijing Zhao
- Department of Mechanical EngineeringNational University of SingaporeSingapore117575Singapore
| | - Xinwei Li
- Faculty of Science, Agriculture, and EngineeringNewcastle UniversitySingapore567739Singapore
| | - Junhao Ding
- Department of Mechanical and Automation EngineeringChinese University of Hong KongSha TinHong Kong999077China
| | - Shuo Qu
- Department of Mechanical and Automation EngineeringChinese University of Hong KongSha TinHong Kong999077China
| | - Quyang Liu
- Department of Mechanical EngineeringNational University of SingaporeSingapore117575Singapore
| | - Zongxin Hu
- Department of Mechanical and Automation EngineeringChinese University of Hong KongSha TinHong Kong999077China
| | - Rui Li
- Department of Mechanical and Automation EngineeringChinese University of Hong KongSha TinHong Kong999077China
| | - Quanqing Tao
- Department of Mechanical and Automation EngineeringChinese University of Hong KongSha TinHong Kong999077China
| | - Haoming Mo
- Department of Mechanical and Automation EngineeringChinese University of Hong KongSha TinHong Kong999077China
| | - Wei Zhai
- Department of Mechanical EngineeringNational University of SingaporeSingapore117575Singapore
| | - Xu Song
- Department of Mechanical and Automation EngineeringChinese University of Hong KongSha TinHong Kong999077China
| |
Collapse
|
36
|
Wang Y, Li Z, Yu R, Chen Y, Wang D, Zhao W, Ge S, Liu H, Li J. Metal-phenolic network biointerface-mediated cell regulation for bone tissue regeneration. Mater Today Bio 2025; 30:101400. [PMID: 39759849 PMCID: PMC11699301 DOI: 10.1016/j.mtbio.2024.101400] [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: 11/07/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Bone tissue regeneration presents a significant challenge in clinical treatment due to inadequate coordination between implant materials and reparative cells at the biomaterial-bone interfaces. This gap underscores the necessity of enhancing interaction modulation between cells and biomaterials, which is a crucial focus in bone tissue engineering. Metal-polyphenolic networks (MPN) are novel inorganic-organic hybrid complexes that are formed through coordination interactions between phenolic ligands and metal ions. These networks provide a multifunctional platform for biomedical applications, with the potential for tailored design and modifications. Despite advances in understanding MPN and their role in bone tissue regeneration, a comprehensive overview of the related mechanisms is lacking. Here, we address this gap by focusing on MPN biointerface-mediated cellular regulatory mechanisms during bone regeneration. We begin by reviewing the natural healing processes of bone defects, followed by a detailed examination of MPN, including their constituents and distinctive characteristics. We then explore the regulatory influence of MPN biointerfaces on key cellular activities during bone regeneration. Additionally, we illustrate their primary applications in addressing inflammatory bone loss, regenerating critical-size bone defects, and enhancing implant-bone integration. In conclusion, this review elucidates how MPN-based interfaces facilitate effective bone tissue regeneration, advancing our understanding of material interface-mediated cellular control and the broader field of tissue engineering.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Zhibang Li
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Ruiqing Yu
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Yi Chen
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Danyang Wang
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Weiwei Zhao
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Shaohua Ge
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, China
| | - Jianhua Li
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| |
Collapse
|
37
|
Xu L, Zhang L, Sun Q, Zhang X, Zhang J, Zhao X, Hu Z, Zhang S, Shi F. Melatonin antagonizes bone loss induced by mechanical unloading via IGF2BP1-dependent m 6A regulation. Cell Mol Life Sci 2025; 82:60. [PMID: 39849105 PMCID: PMC11757843 DOI: 10.1007/s00018-025-05588-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
Disuse bone loss is prone to occur in individuals who lack mechanical stimulation due to prolonged spaceflight or extended bed rest, rendering them susceptible to fractures and placing an enormous burden on social care; nevertheless, the underlying molecular mechanisms of bone loss caused by mechanical unloading have not been fully elucidated. Numerous studies have focused on the epigenetic regulation of disuse bone loss; yet limited research has been conducted on the impact of RNA modification bone formation in response to mechanical unloading conditions. In this study, we discovered that m6A reader IGF2BP1 was downregulated in both osteoblasts treated with 2D clinostat and bone tissue in HLU mice. Supplementing IGF2BP1 could promote osteoblast proliferation and partially alleviate the adverse effects of mechanical unloading on bone formation. Mechanistically, IGF2BP1 inhibited the degradation of Lef1 mRNA by directly binding to its mRNA and recognizing the m6A modification. Furthermore, LEF1 promoted osteoblast proliferation by upregulating c-Myc and Cyclin D1 expression, as well as participated in mediating IGF2BP1-induced osteoblast activity under mechanical unloading. Notably, Melatonin (MT) might participate in the regulation of the IGF2BP1/LEF1 axis, thereby regulating the proliferation of osteoblasts and bone formation. Collectively, this study revealed a new insight into the regulation of the MT/IGF2BP1/LEF1 pathway in the process of unloading-induced bone loss, which could potentially contribute to establishing therapeutic strategies for disuse osteoporosis.
Collapse
Affiliation(s)
- Liqun Xu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Lijun Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, 050081, Hebei, China
| | - Quan Sun
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Xiaoyan Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
- Department of Otolaryngology Head and Neck Surgery, Western Theater Air Force Hospital of PLA, Chengdu, 610065, Sichuan, China
| | - Junfei Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Xiran Zhao
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Zebing Hu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China.
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China.
| | - Fei Shi
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
38
|
Ding Z, Li W, Qi H, Fang T, Zhu Q, Qu X, Chen C, Sun J, Pang Y. The L-shaped association between body roundness index and all-cause mortality in osteoporotic patients: a cohort study based on NHANES data. Front Nutr 2025; 12:1538766. [PMID: 39902313 PMCID: PMC11788163 DOI: 10.3389/fnut.2025.1538766] [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: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 02/05/2025] Open
Abstract
Purpose This study aims to investigate the relationship between the body roundness index (BRI) and overall mortality rates in individuals with osteoporosis (OP), utilizing information sourced from the NHANES database, in order to assess BRI's capability as an indicator for predicting mortality risk. Methods Data from NHANES (2005 to 2010, 2013-2014, and 2017-2018) were analyzed, including 1,596 osteoporotic individuals aged 50 and above. BRI was calculated based on waist circumference (WC) and height, categorizing participants into high (>4.07) and low (≤4.07) BRI groups. To analyze the relationship between BRI and mortality while accounting for important covariates, we employed weighted Cox proportional hazards models, conducted Kaplan-Meier survival analyses, and utilized restricted cubic splines (RCS). Results Higher BRI was significantly associated with better long-term survival, showing an "L"-shaped nonlinear inverse relationship with mortality, with a threshold at BRI = 5. In subgroup analyses, this association remained relatively stable. Conclusion The "L"-shaped association between BRI and mortality indicates that BRI may serve as a useful indicator for evaluating mortality risk in patients with OP, thereby informing clinical interventions and public health approaches.
Collapse
Affiliation(s)
- Ziyao Ding
- First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou, China
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenbo Li
- First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou, China
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Haixu Qi
- First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou, China
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tianci Fang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qirui Zhu
- First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou, China
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xinzhe Qu
- First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou, China
| | - Changchang Chen
- First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jun Sun
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou, China
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yong Pang
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
39
|
Xu YL, Huang M, Zhang Y, Su XY, Huang M, Zou NY, Jiao YR, Sun YC, Liu L, Lei YH, Li CJ. Polycystin-1 regulates tendon-derived mesenchymal stem cells fate and matrix organization in heterotopic ossification. Bone Res 2025; 13:11. [PMID: 39833160 PMCID: PMC11746979 DOI: 10.1038/s41413-024-00392-y] [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: 03/17/2024] [Revised: 09/24/2024] [Accepted: 11/13/2024] [Indexed: 01/22/2025] Open
Abstract
Mechanical stress modulates bone formation and organization of the extracellular matrix (ECM), the interaction of which affects heterotopic ossification (HO). However, the mechanically sensitive cell populations in HO and the underlying mechanism remain elusive. Here, we show that the mechanical protein Polysyctin-1 (PC1, Pkd1) regulates CTSK lineage tendon-derived mesenchymal stem cell (TDMSC) fate and ECM organization, thus affecting HO progression. First, we revealed that CTSK lineage TDMSCs are the major source of osteoblasts and fibroblasts in HO and are responsive to mechanical cues via single-cell RNA sequencing analysis and experiments with a lineage tracing mouse model. Moreover, we showed that PC1 mediates the mechanosignal transduction of CTSK lineage TDMSCs to regulate osteogenic and fibrogenic differentiation and alters the ECM architecture by facilitating TAZ nuclear translocation. Conditional gene depletion of Pkd1 or Taz in CTSK lineage cells and pharmaceutical intervention in the PC1-TAZ axis disrupt osteogenesis, fibrogenesis and ECM organization, and consequently attenuate HO progression. These findings suggest that mechanically sensitive CTSK-lineage TDMSCs contribute to heterotopic ossification through PC1-TAZ signaling axis mediated cell fate determination and ECM organization.
Collapse
Affiliation(s)
- Yi Li Xu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- Department of Orthodontics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yang Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan Province, 410013, China
| | - Xin Ying Su
- Department of Orthodontics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Min Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Nan Yu Zou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu Rui Jiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu Chen Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Ling Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yong Hua Lei
- Department of Orthodontics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Chang Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China.
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Laboratory Animal Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
40
|
Holzer CS, Pukaluk A, Viertler C, Regitnig P, Charry EM, Wolinski H, Eschbach M, Caulk AW, Holzapfel GA. Implications of compressive loading of the stomach wall: Interplay between mechanical deformation and microstructure. Acta Biomater 2025; 192:101-118. [PMID: 39694163 DOI: 10.1016/j.actbio.2024.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
During gastric surgery, the stomach wall is compressed with clamps and sutures or staple lines. These short- and long-term deformations can severely compromise the integrity of the tissue and make it difficult for the stomach wall to respond and remodel to the new loading conditions. Consequently, serious intra- and postoperative complications such as the formation of leaks during bariatric surgeries, can be associated with these immense tissue deformations. Hence, the study aimed to investigate the effects of compressive loading of the stomach wall in the radial direction. This was done by macroscopic mechanical loading of the stomach wall in each region of the stomach and evaluating the microstructural changes inflicted in the tissue. For this purpose, several imaging techniques were used, i.e., a histological analysis, second-harmonic generation microscopy, and X-ray micro-computed tomography. The combination of these three methods allowed us to investigate the gradual compression of the different stomach layers as well as the local reorientation and deformation of the main microstructural components, e.g., collagen fibers and muscle bundles. Importantly, this study found that the collagen bundles in the stomach wall straighten and reorient toward the circumferential-longitudinal plane and partially fan out with increased radial compressive deformation. The 3D scans of the stomach wall indicated a deterioration of the blood vessels and buckling of the mucosal glands due to compression. Statement of significance Unfortunately, little is known about the load transfer in the stomach wall during gastric surgery and the associated deformations on the macro- and microscale. The present study investigates the structural changes of the stomach wall, its layers and the inherent biological building blocks using histology, multi-photon microscopy, and micro-computed tomography. For the first time, the layer-specific response to stepwise radial compression of the stomach wall was studied, the related collagen fiber parameters were estimated, and a 3D sample structure was visualized. This clinically-oriented study links the structural changes within the wall to the postoperative remodel- ing process and the irreversibly altered gastric motility, thereby underscoring its relevance to the field of biomedical engineering, e.g., the development and improvement of surgical instruments.
Collapse
Affiliation(s)
| | - Anna Pukaluk
- Institute of Biomechanics, Graz University of Technology, Austria
| | - Christian Viertler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Austria
| | - Peter Regitnig
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Austria
| | | | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Austria; Field of Excellence BioHealth - University of Graz, Austria
| | | | | | - Gerhard A Holzapfel
- Institute of Biomechanics, Graz University of Technology, Austria; Department of Structural Engineering, NTNU, Norway.
| |
Collapse
|
41
|
Quan S, Yang J, Huang S, Shao J, Liu Y, Yang H. Silk fibroin as a potential candidate for bone tissue engineering applications. Biomater Sci 2025; 13:364-378. [PMID: 39620282 DOI: 10.1039/d4bm00950a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Silk fibroin (SF), a pivotal biomaterial, holds immense promise for diverse applications within the realm of bone tissue engineering. SF is an ideal scaffold material with exceptional biocompatibility, mechanical robustness, biodegradability, and bioactivity. A plethora of investigations have corroborated SF's efficacy in supporting bone tissue repair and regeneration. This comprehensive review delves into the structural attributes, physicochemical characteristics, and extraction methodologies of SF. Moreover, it elucidates the strides taken in harnessing SF across a spectrum of forms, including films, hydrogels, scaffolds, electrospun fibers, and composites for bone tissue engineering applications. Moreover, the application bottleneck of SF as a bone repair material is highlighted, and its development prospects and potential biomedical applications are also presented in this review. We expect that this review can inspire the broad interest of a wide range of readers working in the fields of materials science, tissue engineering, biomaterials, bioengineering, and biomedicine.
Collapse
Affiliation(s)
- Shaohao Quan
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China.
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510182, China.
| | - Jie Yang
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China.
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Sirui Huang
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Jundong Shao
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510182, China.
| | - Yang Liu
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213000, China.
| | - Hui Yang
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China.
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
42
|
Wang LY, Liang LM, Zhang XX, Chi H, Peng FL. Short bouts and long-term exercise reduce sedentary-induced bone loss and microstructural changes by modulating bone formation and resorption in healthy young male rats. Sci Rep 2025; 15:1825. [PMID: 39805876 PMCID: PMC11730605 DOI: 10.1038/s41598-024-82243-8] [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: 06/23/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
Although the toxic effect of Sedentary behavior (SED) on bone health has been demonstrated in the previous study, the underlying mechanisms of SED, or break SED to bone health remain unclear. In this study, we aim to investigate the effects of sedentary behavior (SED) on bone health, as well as the potential favor effects of moderate to vigorous physical activity (MVPA) and periodic interruptions of SED. To simulate SED, we used small Plexiglas cages (20.0 × 9.0 × 10.0 cm) to restrict animal movement. Short bursts of exercise to break SED and continuous long-term exercise were also designed. After an 8-weeks period of SED, we observed decreased bone mass and bone microstructure. Specifically, there was a notable decrease in the bone mineral density (BMD), bone surface (BS) and cortical thickness (Ct.Th) significantly reduced in cortical bone. In the trabecular bone, parameters such as trabecular separation (Tb.Sp), trabecular number (Tb.N), BS, connectivity density (Conn.D), BS/BV, bone volume/tissue volume (BV/TV), degree of anisotropy (DA), and structural model index (SMI) were also significantly reduced. In addition, we detected an increase in serum tartrate-resistant acid phosphatase (TRAP) levels in SED rats at both 4 and 8 weeks. At 8 weeks, the osteoclast number and surface with TRAP-staining were significantly increased, however, the OPG mRNA and proteins level were significantly decreased. After daily short bouts exercise and long-term exercise, we observed improvements in bone mass and microstructure. These improvements included increasing BMD and BV/TV of cortical bone, and improving Conn.D, BV/TV, DA and SMI of trabecular. Meanwhile, we found that, at 4 and 8 weeks, there was an increase in serum ALP. At 8 weeks, the mineralized nodules surface with Alizarin Red S-staining, and OPG mRNA and proteins level in bone tissue were significantly increased. Our findings suggest that SED leads to alterations in the bone mass and microstructure, which are associated with the changes in the OPG protein and bone remodeling. Exercise, whether in short daily bouts or continuous long-term sessions, can ameliorate the harmful effects of SED. Similarly, the changes in bone mass and microstructure from exercise are also associated with the changes in the OPG protein and bone remodeling by upregulated osteoblast activity to bone formation. Overall, our findings indicate the importance of physical activity in maintaining bone health and preventing the negative impacts of prolonged SED.
Collapse
Affiliation(s)
- L Y Wang
- Guangxi Normal University, Guangxi, China
- Guangxi Medical University, Guangxi, China
| | - L M Liang
- Guangxi Normal University, Guangxi, China.
| | - X X Zhang
- Guangxi Normal University, Guangxi, China
| | - H Chi
- Guangxi Normal University, Guangxi, China
| | - F L Peng
- Guangxi Normal University, Guangxi, China.
| |
Collapse
|
43
|
Le Grill S, Brouillet F, Drouet C. Bone Regeneration: Mini-Review and Appealing Perspectives. Bioengineering (Basel) 2025; 12:38. [PMID: 39851312 PMCID: PMC11763268 DOI: 10.3390/bioengineering12010038] [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: 10/30/2024] [Revised: 12/18/2024] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
Bone is a natural mineral-organic nanocomposite protecting internal organs and allowing mobility. Through the ages, numerous strategies have been developed for repairing bone defects and fixing fractures. Several generations of bone repair biomaterials have been proposed, either based on metals, ceramics, glasses, or polymers, depending on the clinical need, the maturity of technologies, and knowledge of the natural constitution of the bone tissue to be repaired. The global trend in bone implant research is shifting toward osteointegrative, bioactive and possibly stimuli-responsive biomaterials and, where possible, resorbable implants that actively promote the regeneration of natural bone tissue. In this mini-review, the fundamentals of bone healing materials and clinical challenges are summarized and commented on with regard to progressing scientific discoveries. The main types of bone-healing materials are then reviewed, and their specific relevance to the field is reminded, with the citation of reference works. In the final part, we highlight the promise of hybrid organic-inorganic bioactive materials and the ongoing research activities toward the development of multifunctional or stimuli-responsive implants. This contribution is expected to serve as a commented introduction to the ever-progressing field of bone regeneration and highlight trends of future-oriented research.
Collapse
Affiliation(s)
- Sylvain Le Grill
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, 4 Allée Emile Monso, BP44362, CEDEX 4, 31030 Toulouse, France; (F.B.); (C.D.)
- Regenerative Nanomedicine Unit, Center of Research on Biomedicines of Strasbourg (CRBS), French National Institute of Health and Medical Research (INSERM), University of Strasbourg, UMR 1260, 1 Rue Eugène Boeckel, 67000 Strasbourg, France
| | - Fabien Brouillet
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, 4 Allée Emile Monso, BP44362, CEDEX 4, 31030 Toulouse, France; (F.B.); (C.D.)
| | - Christophe Drouet
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, 4 Allée Emile Monso, BP44362, CEDEX 4, 31030 Toulouse, France; (F.B.); (C.D.)
| |
Collapse
|
44
|
Park H, Trupiano SP, Medarev SL, Ghosh P, Caldwell JT, Yarrow JF, Muller-Delp JM. Aerobic exercise training-induced bone and vascular adaptations in mice lacking adiponectin. Bone 2025; 190:117272. [PMID: 39369833 PMCID: PMC11795456 DOI: 10.1016/j.bone.2024.117272] [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: 07/29/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Adiponectin regulates lipid and glucose metabolism, and insulin sensitivity in various target organs; however, the effects of adiponectin on bone health remain controversial. Exercise training can enhance bone density, bone microarchitecture, and blood flow. This study aimed to elucidate the role of adiponectin in adaptations of bone microarchitecture and bone vasculature in response to aerobic exercise training. Adult male C57BL/6 wild-type (WT) and homozygous adiponectin knockout (AdipoKO) mice were either treadmill exercise trained or remained sedentary for 8-10 weeks. The trabecular structures of the distal femoral metaphysis, a weight-bearing bone, and the mandible, a non-weight-bearing bone, were examined using microcomputed tomography. The femoral principal nutrient arteries were isolated to assess vasoreactivity (vasodilation and vasoconstriction) and structural remodeling. At the femoral metaphysis, impaired trabecular bone structures, including reduced connectivity density and increased trabecular spacing, were observed in AdipoKO mice compared to WT mice. In addition, nitric oxide-mediated, endothelium-dependent vasodilation was substantially reduced, and wall-to-lumen ratio was significantly increased in the femoral principal nutrient artery of AdipoKO mice. Interestingly, although exercise training-induced enhancements in trabecular connectivity density were observed at the femoral metaphysis of both WT and AdipoKO, increased vasoconstrictor responses were only observed in the femoral principal nutrient artery of WT mice, not in the AdipoKO mice. In mandibular trabecular bone, exercise training increased trabecular bone volume fraction (BV/TV, %) and intersection surface in the mandible of both WT and AdipoKO mice. These findings indicate that adiponectin is crucial for maintaining normal bone microarchitecture and vasculature. Although the absence of adiponectin compromises bone vascular adaptation to exercise training in mice, some exercise training-induced alterations in bone microarchitecture occurred in the absence of adiponectin, suggesting contribution of compensatory mechanisms during exercise training.
Collapse
Affiliation(s)
- Hyerim Park
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Samuel P Trupiano
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Steven L Medarev
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Payal Ghosh
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Jacob T Caldwell
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA; Department of Exercise and Sport Science, University of Wisconsin-La Crosse, La Crosse, WI, USA
| | - Joshua F Yarrow
- Eastern Colorado Geriatrics Research, Education, and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO, USA
| | - Judy M Muller-Delp
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
45
|
Wang Y, Yung P, Lu G, Liu Y, Ding C, Mao C, Li ZA, Tuan RS. Musculoskeletal Organs-on-Chips: An Emerging Platform for Studying the Nanotechnology-Biology Interface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2401334. [PMID: 38491868 PMCID: PMC11733728 DOI: 10.1002/adma.202401334] [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: 01/25/2024] [Revised: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Nanotechnology-based approaches are promising for the treatment of musculoskeletal (MSK) disorders, which present significant clinical burdens and challenges, but their clinical translation requires a deep understanding of the complex interplay between nanotechnology and MSK biology. Organ-on-a-chip (OoC) systems have emerged as an innovative and versatile microphysiological platform to replicate the dynamics of tissue microenvironment for studying nanotechnology-biology interactions. This review first covers recent advances and applications of MSK OoCs and their ability to mimic the biophysical and biochemical stimuli encountered by MSK tissues. Next, by integrating nanotechnology into MSK OoCs, cellular responses and tissue behaviors may be investigated by precisely controlling and manipulating the nanoscale environment. Analysis of MSK disease mechanisms, particularly bone, joint, and muscle tissue degeneration, and drug screening and development of personalized medicine may be greatly facilitated using MSK OoCs. Finally, future challenges and directions are outlined for the field, including advanced sensing technologies, integration of immune-active components, and enhancement of biomimetic functionality. By highlighting the emerging applications of MSK OoCs, this review aims to advance the understanding of the intricate nanotechnology-MSK biology interface and its significance in MSK disease management, and the development of innovative and personalized therapeutic and interventional strategies.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Patrick Yung
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Gang Lu
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Yuwei Liu
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- The First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhenGuangdong518037P. R. China
| | - Changhai Ding
- Clinical Research CentreZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510260China
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Zhong Alan Li
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Key Laboratory of Regenerative MedicineMinistry of EducationSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077P. R. China
- Shenzhen Research InstituteThe Chinese University of Hong KongShenzhen518172P. R. China
| | - Rocky S. Tuan
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| |
Collapse
|
46
|
Randhawa A, Ganguly K, Dutta SD, Patil TV, Lim KT. Transcriptomic profiling of human mesenchymal stem cells using a pulsed electromagnetic-wave motion bioreactor system for enhanced osteogenic commitment and therapeutic potentials. Biomaterials 2025; 312:122713. [PMID: 39084096 DOI: 10.1016/j.biomaterials.2024.122713] [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: 02/13/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Traditional bioreactor systems involve the use of three-dimensional (3D) scaffolds or stem cell aggregates, limiting the accessibility to the production of cell-secreted biomolecules. Herein, we present the use a pulse electromagnetic fields (pEMFs)-assisted wave-motion bioreactor system for the dynamic and scalable culture of human bone marrow-derived mesenchymal stem cells (hBMSCs) with enhanced the secretion of various soluble factors with massive therapeutic potential. The present study investigated the influence of dynamic pEMF (D-pEMF) on the kinetic of hBMSCs. A 30-min exposure of pEMF (10V-1Hz, 5.82 G) with 35 oscillations per minute (OPM) rocking speed can induce the proliferation (1 × 105 → 4.5 × 105) of hBMSCs than static culture. Furthermore, the culture of hBMSCs in osteo-induction media revealed a greater enhancement of osteogenic transcription factors under the D-pEMF condition, suggesting that D-pEMF addition significantly boosted hBMSCs osteogenesis. Additionally, the RNA sequencing data revealed a significant shift in various osteogenic and signaling genes in the D-pEMF group, further suggesting their osteogenic capabilities. In this research, we demonstrated that the combined effect of wave and pEMF stimulation on hBMSCs allows rapid proliferation and induces osteogenic properties in the cells. Moreover, our study revealed that D-pEMF stimuli also induce ROS-scavenging properties in the cultured cells. This study also revealed a bioactive and cost-effective approach that enables the use of cells without using any expensive materials and avoids the possible risks associated with them post-implantation.
Collapse
Affiliation(s)
- Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea; Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
47
|
Gamble JG. Pediatric Fracture Remodeling: From Wolff to Wnt. Cureus 2025; 17:e78266. [PMID: 39897217 PMCID: PMC11782688 DOI: 10.7759/cureus.78266] [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] [Accepted: 01/30/2025] [Indexed: 02/04/2025] Open
Abstract
Pediatric fracture remodeling is a complex mechanobiological process in which a team of cells, including osteoclasts, osteoblasts, and osteocytes, responds to cytokine and mechanical signals to synthesize new bone in areas of high stress (concavity of a fracture) and remove older redundant bone in areas of low stress (convexity and medullary canal). Piezo1 mechanoreceptors and other pressure-sensitive membrane proteins perceive and convert mechanical strains into intracellular chemical signals. Cytokines are peptides that bind to cell membrane receptors and influence cell functions. Bone morphogenetic proteins and Wnt are the major osteogenic cytokines. Macrophage colony-stimulating factor and receptor activator of nuclear factor κB ligand (RANKL) are the major osteoclastic cytokines. The combination of mechanical stresses and cytokine concentrations stimulates osteoclasts to resorb bone and osteoblasts to make new bone, resulting in remodeling that restores bone strength and structure.
Collapse
Affiliation(s)
- James G Gamble
- Orthopaedic Surgery, Stanford University School of Medicine, Lucile Packard Children's Hospital, Stanford, USA
| |
Collapse
|
48
|
Alontseva D, (Yantsen) YS, Voinarovych S, Obrosov A, Yamanoglu R, Khoshnaw F, Nessipbekova A, Syzdykova A, Yavuz HI, Kaliuzhnyi S, Krasavin A, Azamatov B, Khozhanov A, Olzhayev F, Weiß S. Microplasma-Sprayed Titanium and Hydroxyapatite Coatings on Ti6Al4V Alloy: in vitro Biocompatibility and Corrosion Resistance: Part I. JOHNSON MATTHEY TECHNOLOGY REVIEW 2025; 69:45-58. [DOI: 10.1595/205651325x17201903387613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
This two-part paper investigates the bioactivity and mechanical properties of coatings applied to Ti6Al4V, a common titanium alloy used in endoprosthetic implants. Coatings made from hydroxyapatite (HA) powder and commercially pure titanium (CP-Ti) wires were applied using microplasma spraying. The study focuses on the responses of rat mesenchymal stem cells (MSCs), which are essential for bone healing, to these coatings. Part I shows how adjusting the microplasma spraying process allows coatings with varying porosity and surface roughness to be achieved.
Collapse
Affiliation(s)
- Darya Alontseva
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000 Astana, Kazakhstan; School of Digital Technologies and Artificial Intelligence, D. Serikbayev East Kazakhstan Technical University, 19 Serikbayev Street, 070010, Ust-Kamenogorsk, Kazakhstan
| | - Yuliya Safarova (Yantsen)
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000, Astana, Kazakhstan
| | - Sergii Voinarovych
- E.O. Paton Electric Welding Institute of NAS of Ukraine, 11 Kazymyr Malevich Street, 03150, Kyiv, Ukraine
| | - Aleksei Obrosov
- Department of Physical Metallurgy and Materials Technology, Brandenburg Technical University, Cottbus 03046, Germany
| | - Ridvan Yamanoglu
- Department of Metallurgical and Materials Engineering, Faculty of Engineering, Kocaeli University, 41001, Kocaeli, Türkiye
| | - Fuad Khoshnaw
- School of Engineering and Sustainable Development, Faculty of Computing, Engineering and Media, De Montfort University, LE1 9BH, Leicester, UK
| | - Assem Nessipbekova
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000, Astana, Kazakhstan
| | - Aizhan Syzdykova
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000, Astana, Kazakhstan
| | - Hasan Ismail Yavuz
- Department of Metallurgical and Materials Engineering, Faculty of Engineering, Kocaeli University, 41001, Kocaeli, Türkiye
| | - Sergii Kaliuzhnyi
- E.O. Paton Electric Welding Institute of NAS of Ukraine, 11 Kazymyr Malevich Street, 03150, Kyiv, Ukraine
| | - Alexander Krasavin
- School of Digital Technologies and Artificial Intelligence, D. Serikbayev East Kazakhstan Technical University, 19 Serikbayev Street, 070010, Ust-Kamenogorsk, Kazakhstan
| | - Bagdat Azamatov
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000 Astana, Kazakhstan; Smart Engineering Competence Centre, D. Serikbayev East Kazakhstan Technical University, 19 Serikbayev Street, 070010, Ust-Kamenogorsk, Kazakhstan
| | - Alexandr Khozhanov
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000 Astana, Kazakhstan; Smart Engineering Competence Centre, D. Serikbayev East Kazakhstan Technical University, 19 Serikbayev Street, 070010, Ust-Kamenogorsk, Kazakhstan
| | - Farkhad Olzhayev
- Laboratory of Bioengineering and Regenerative Medicine, National Laboratory Astana, Nazarbayev University, 010000, Astana, Kazakhstan
| | - Sabine Weiß
- Department of Physical Metallurgy and Materials Technology, Brandenburg Technical University, Cottbus 03046, Germany
| |
Collapse
|
49
|
Dalle Carbonare L, Braggio M, Minoia A, Cominacini M, Romanelli MG, Pessoa J, Tiso N, Valenti MT. Modeling Musculoskeletal Disorders in Zebrafish: Advancements in Muscle and Bone Research. Cells 2024; 14:28. [PMID: 39791729 PMCID: PMC11719663 DOI: 10.3390/cells14010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025] Open
Abstract
Zebrafish (Danio rerio) have emerged as a valuable model organism for investigating musculoskeletal development and the pathophysiology of associated diseases. Key genes and biological processes in zebrafish that closely mirror those in humans, rapid development, and transparent embryos make zebrafish ideal for the in vivo studies of bone and muscle formation, as well as the molecular mechanisms underlying musculoskeletal disorders. This review focuses on the utility of zebrafish in modeling various musculoskeletal conditions, with an emphasis on bone diseases such as osteoporosis and osteogenesis imperfecta, as well as muscle disorders like Duchenne muscular dystrophy. These models have provided significant insights into the molecular pathways involved in these diseases, helping to identify the key genetic and biochemical factors that contribute to their progression. These findings have also advanced our understanding of disease mechanisms and facilitated the development of potential therapeutic strategies for musculoskeletal disorders.
Collapse
Affiliation(s)
- Luca Dalle Carbonare
- Department of Engineering for the Innovation Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.C.)
| | - Michele Braggio
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (M.B.); (M.G.R.)
| | - Arianna Minoia
- Department of Engineering for the Innovation Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.C.)
| | - Mattia Cominacini
- Department of Engineering for the Innovation Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.C.)
| | - Maria Grazia Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (M.B.); (M.G.R.)
| | - João Pessoa
- Department of Medical Sciences and Institute of Biomedicine—iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Natascia Tiso
- Department of Biology, University of Padua, 35131 Padua, Italy;
| | - Maria Teresa Valenti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (M.B.); (M.G.R.)
| |
Collapse
|
50
|
Hia EM, Suh IW, Jang SR, Park CH. Magnetically responsive micro-clustered calcium phosphate-reinforced cell-laden microbead sodium alginate hydrogel for accelerated osteogenic tissue regeneration. Carbohydr Polym 2024; 346:122666. [PMID: 39245476 DOI: 10.1016/j.carbpol.2024.122666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/18/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
The rising prevalence of bone injuries has increased the demand for minimally invasive treatments. Microbead hydrogels, renowned for cell encapsulation, provide a versatile substrate for bone tissue regeneration. They deliver bioactive agents, support cell growth, and promote osteogenesis, aiding bone repair and regeneration. In this study, we synthesized superparamagnetic iron oxide nanoparticles (Sp) coated with a calcium phosphate layer (m-Sp), achieving a distinctive flower-like micro-cluster morphology. Subsequently, sodium alginate (SA) microbead hydrogels containing m-Sp (McSa@m-Sp) were fabricated using a dropping gelation strategy. McSa@m-Sp is magnetically targetable, enhance cross-linking, control degradation rates, and provide strong antibacterial activity. Encapsulation studies with MC3T3-E1 cells revealed enhanced viability and proliferation. These studies also indicated significantly elevated alkaline phosphatase (ALP) activity and mineralization in MC3T3-E1 cells, as confirmed by Alizarin Red S (ARS) and Von Kossa staining, along with increased collagen production within the McSa@m-Sp microbead hydrogels. Immunocytochemistry (ICC) and gene expression studies supported the osteoinductive potential of McSa@m-Sp, showing increased expression of osteogenic markers including RUNX-2, collagen-I, osteopontin, and osteocalcin. Thus, McSa@m-Sp microbead hydrogels offer a promising strategy for multifunctional scaffolds in bone tissue engineering.
Collapse
Affiliation(s)
- Esensil Man Hia
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea; Department of Bionanotechnology and Bioconvergence Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea
| | - Il Won Suh
- Department of Bionanotechnology and Bioconvergence Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea
| | - Se Rim Jang
- Division of Mechanical Design Engineering, Jeonbuk National University, Jeonju 561-756, Republic of Korea
| | - Chan Hee Park
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea; Department of Bionanotechnology and Bioconvergence Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea; Division of Mechanical Design Engineering, Jeonbuk National University, Jeonju 561-756, Republic of Korea.
| |
Collapse
|