1
|
Findeisen L, Tian X, Vater C, Raina DB, Kern H, Bolte J, Straßburger L, Matuszewski LM, Modler N, Gottwald R, Winkler A, Schaser KD, Disch AC, Zwingenberger S. Exploring an innovative augmentation strategy in spinal fusion: A novel selective prostaglandin EP4 receptor agonist as a potential osteopromotive factor to enhance lumbar posterolateral fusion. Biomaterials 2025; 320:123278. [PMID: 40132358 DOI: 10.1016/j.biomaterials.2025.123278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND On-site delivery of bioactive agents facilitates enhancing the effectiveness of spinal fusion. However, the FDA-approved agents currently used in clinical practice are limited by side effects and cost issues, urging exploration of new alternatives. AIM This study aimed to investigate the effectiveness of KMN-159, a novel selective prostaglandin EP4 receptor agonist with osteopromotive properties, in spinal posterolateral fusion (PLF) surgery. METHODS Various doses of KMN-159 were delivered locally using a mineralized collagen matrix (MCM) scaffold, and its efficacy results were compared with FDA-approved recombinant human bone morphogenetic protein-2 (rhBMP-2) in a rat lumbar PLF model. 192 male Wistar rats, aged 10 weeks, were randomized into 8 groups: 1) SHAM, 2) MCM, 3) MCM +10 μg rhBMP-2 (per scaffold), 4-8) MCM + 0.1, 1, 10, 100 or 1000 μg KMN-159 (per scaffold). PLF surgery was performed at the L4-5 level, and animals were euthanized after 3 and 6 weeks for spinal fusion evaluation. RESULTS KMN-159 exhibited dose-dependent osteopromotive effects on osteoblasts, osteoclasts, and vascular ingrowth within MCM carriers, resulting in new bone formation in a dose-dependent manner. The mid- and high-dose KMN-159 (10, 100, and 1000 μg) groups significantly enhanced PLF with biomechanical improvement, while low-dose (0.1 and 1 μg) groups were insufficient to achieve lumbar fusion. CONCLUSION KMN-159 emerges as a novel osteopromotive factor, coupled with its functionalized MCM scaffold presents a potential bioactive material for enhancing PLF surgery outcomes.
Collapse
Affiliation(s)
- Lisa Findeisen
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany
| | - Xinggui Tian
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany.
| | - Corina Vater
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany
| | - Deepak Bushan Raina
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Lund, 22185, Sweden
| | - Hannes Kern
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany
| | - Julia Bolte
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany
| | - Luisa Straßburger
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany
| | - Lucas-Maximilian Matuszewski
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany
| | - Niels Modler
- Institute of Lightweight Engineering and Polymer Technology at TUD Dresden University of Technology, 01062, Dresden, Germany
| | - Robert Gottwald
- Institute of Lightweight Engineering and Polymer Technology at TUD Dresden University of Technology, 01062, Dresden, Germany
| | - Anja Winkler
- Institute of Lightweight Engineering and Polymer Technology at TUD Dresden University of Technology, 01062, Dresden, Germany
| | - Klaus-Dieter Schaser
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany
| | - Alexander C Disch
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany
| | - Stefan Zwingenberger
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany; Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine at TUD Dresden University of Technology, 01307, Dresden, Germany
| |
Collapse
|
2
|
Yu W, Hu L, Wei Y, Xue C, Liu Y, Xie H. Advances of novel hydrogels in the healing process of alveolar sockets. BIOMATERIALS ADVANCES 2025; 173:214280. [PMID: 40086007 DOI: 10.1016/j.bioadv.2025.214280] [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: 01/13/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Tooth extraction is a common oral surgical procedure that often leads to delayed alveolar socket healing due to the complexity of the oral microenvironment, which can hinder the patient's aesthetic and functional recovery. Effective alveolar socket healing requires a multidisciplinary approach. Recent advancements in materials science and bioengineering have facilitated the development of innovative strategies, with hydrogels emerging as ideal restorative materials for alveolar socket repair due to their superior properties. This review provides an overview of recent advances in hydrogels for alveolar socket healing, focusing on their classification, physical properties (e.g., mechanical strength, swelling behavior, degradation rate, and injectability), biological functions, and applications in relevant animal models. Specifically, the bone-regenerative and antimicrobial properties of hydrogels are highlighted. Furthermore, this review identifies future directions and addresses challenges associated with the clinical application of hydrogels in extraction socket healing.
Collapse
Affiliation(s)
- Wenqing Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Liwei Hu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Yige Wei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Chengyu Xue
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Yunfei Liu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, PR China.
| | - Huixu Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
3
|
Mammadov J, Davudov M, Aliyev T, Isgandarova S. The Effect of Immunocorrection on Reparative Osteogenesis in Mandibular Fracture: A Histomorphometric Study. J Craniofac Surg 2025:00001665-990000000-02611. [PMID: 40202323 DOI: 10.1097/scs.0000000000011404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Fracture consolidation is a fundamental factor in understanding the biological process of bone healing in human. The objective of this study was to evaluate the effects of immunocorrection after trauma surgery on bone healing process in mandibular fracture in rabbits. METHODS This study carried out using 24 rabbits. In all rabbits, the mandibular fracture model and osteogenesis carried out on the fracture site by titanium miniplates and screws. Blood analysis performed before and during the treatment. The animals slaughtered and fractured site removed for morphologic studies at baseline and follow-up assessments. RESULTS After surgical trauma in all animals, immunologic indicators of blood including circulating immunocomplex, complements, and lysosomes have been reduced. CONCLUSION General immunity and histomorphometric evaluation of the present study showed posttraumatic immunodeficiency could affects bone healing.
Collapse
Affiliation(s)
- Jahid Mammadov
- Department of Oral and Maxillofacial Surgery, Azerbaijan Medical University, Baku, Azerbaijan
| | | | | | | |
Collapse
|
4
|
Li S, Cai X, Guo J, Li X, Li W, Liu Y, Qi M. Cell communication and relevant signaling pathways in osteogenesis-angiogenesis coupling. Bone Res 2025; 13:45. [PMID: 40195313 PMCID: PMC11977258 DOI: 10.1038/s41413-025-00417-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
Osteogenesis is the process of bone formation mediated by the osteoblasts, participating in various bone-related physiological processes including bone development, bone homeostasis and fracture healing. It exhibits temporal and spatial interconnectivity with angiogenesis, constructed by multiple forms of cell communication occurring between bone and vascular endothelial cells. Molecular regulation among different cell types is crucial for coordinating osteogenesis and angiogenesis to facilitate bone remodeling, fracture healing, and other bone-related processes. The transmission of signaling molecules and the activation of their corresponding signal pathways are indispensable for various forms of cell communication. This communication acts as a "bridge" in coupling osteogenesis to angiogenesis. This article reviews the modes and processes of cell communication in osteogenesis-angiogenesis coupling over the past decade, mainly focusing on interactions among bone-related cells and vascular endothelial cells to provide insights into the mechanism of cell communication of osteogenesis-angiogenesis coupling in different bone-related contexts. Moreover, clinical relevance and applications are also introduced in this review.
Collapse
Affiliation(s)
- Shuqing Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Xinjia Cai
- Central Laboratory, Peking University School and Hospital for Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Jiahe Guo
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Xiaolu Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Wen Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yan Liu
- Central Laboratory, Peking University School and Hospital for Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Mengchun Qi
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China.
| |
Collapse
|
5
|
Zhou W, Liu Y, Nie X, Zhu C, Xiong L, Zhou J, Huang W. Peptide-based inflammation-responsive implant coating sequentially regulates bone regeneration to enhance interfacial osseointegration. Nat Commun 2025; 16:3283. [PMID: 40189598 PMCID: PMC11973180 DOI: 10.1038/s41467-025-58444-8] [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: 09/22/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Aseptic loosening is the primary cause of bone prosthesis failure, commonly attributed to inadequate osseointegration due to coatings misaligned with bone regeneration. Here, we modify the titanium surface with a mussel-inspired peptide to form a 3,4-dihydroxyphenylalanine (DOPA)-rich coating, then graft N3-K15-PVGLIG-K23 (P1) and N3-Y5-PVGLIG-K23 (P2), which are composed of anti-inflammatory (K23), angiogenic (K15), osteogenic (Y5), and inflammation-responsive (PVGLIG) sequences, onto the surface via click chemistry, forming the DOPA-P1@P2 coating. DOPA-P1@P2 promotes bone regeneration through sequential regulation. In the initial stage, the outermost K23 induces M2 macrophage polarization, establishing a pro-regenerative immune microenvironment. Subsequently, K15 and Y5, exposed by the release of K23, enhance angiogenesis and osteogenesis. In the final stage, DOPA-P1@P2 outperforms the TiO₂ control, showing a 161% increase in maximal push-out force, a 207% increase in bone volume fraction, and a 1409% increase in bone-to-implant contact. These findings show that DOPA-P1@P2 efficiently enhances interfacial osseointegration by sequentially regulating bone regeneration, providing viable insights into coating design.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Liu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xuan Nie
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China
| | - Chen Zhu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Liming Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jing Zhou
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
6
|
Maemura M, Morita M, Ogata S, Miyamoto Y, Ida T, Shibusaka K, Negishi S, Hosonuma M, Saito T, Yoshitake J, Takata T, Matsunaga T, Mishima E, Barayeu U, Akaike T, Yano F. Supersulfides contribute to joint homeostasis and bone regeneration. Redox Biol 2025; 81:103545. [PMID: 39983344 PMCID: PMC11893308 DOI: 10.1016/j.redox.2025.103545] [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: 12/04/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
The physiological functions of supersulfides, inorganic and organic sulfides with sulfur catenation, have been extensively studied. Their synthesis is mainly mediated by mitochondrial cysteinyl-tRNA synthetase (CARS2) that functions as a principal cysteine persulfide synthase. This study aimed to investigate the role of supersulfides in joint homeostasis and bone regeneration. Using Cars2AINK/+ mutant mice, in which the KIIK motif of CARS2 essential for supersulfide production was replaced with AINK, we evaluated the role of supersulfides in fracture healing and cartilage homeostasis during osteoarthritis (OA). Tibial fracture surgery was performed on the wild-type (Cars2+/+) and Cars2AINK/+ mice littermates. Bulk RNA-seq analysis for the osteochondral regeneration in the fracture model showed increased inflammatory markers and reduced osteogenic factors, indicative of impaired bone regeneration, in Cars2AINK/+ mice. Destabilization of the medial meniscus (DMM) surgery was performed to produce the mouse OA model. Histological analyses with Osteoarthritis Research Society International and synovitis scores revealed accelerated OA progression in Cars2AINK/+ mice compared with that in Cars2+/+ mice. To assess the effects of supersulfides on OA progression, glutathione trisulfide (GSSSG) or saline was periodically injected into the mouse knee joints after the DMM surgery. Thus, supersulfides derived from CARS2 and GSSSG exogenously administered significantly inhibited inflammation and lipid peroxidation of the joint cartilage, possibly through suppression of ferroptosis, during OA development. This study represents a significant advancement in understanding anti-inflammatory and anti-oxidant functions of supersulfides in skeletal tissues and may have a clinical relevance for the bone healing and OA therapeutics.
Collapse
Affiliation(s)
- Miki Maemura
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Seiryo Ogata
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Yoichi Miyamoto
- Faculty of Arts and Sciences at Fujiyoshida, Showa University, Fujiyoshida, Japan
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Kazuhiro Shibusaka
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan; Department of Orthodontics, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Soichiro Negishi
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Masahiro Hosonuma
- Department of Pharmacology, Graduate School of Pharmacy, Showa University, Tokyo, Japan
| | - Taku Saito
- Sensory & Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Yoshitake
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Tsuyoshi Takata
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Tetsuro Matsunaga
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan; Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita, Japan
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | | | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan.
| | - Fumiko Yano
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan.
| |
Collapse
|
7
|
Ganko R, Madhavan A, Hamouda W, Muthu S, Jain A, Yoon ST, El-Rozz H, Cyril D, Pabbruwe M, Tipper JL, Tavakoli J. Spinal implant wear particles: Generation, characterization, biological impacts, and future considerations. iScience 2025; 28:112193. [DOI: 10.1016/j.isci.2025.112193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
|
8
|
Yu H, Mu Q, Wang Z, Guo Y, Zhao J, Wang G, Wang Q, Meng X, Dong X, Wang S, Sun J. A study on early diagnosis for fracture non-union prediction using deep learning and bone morphometric parameters. Front Med (Lausanne) 2025; 12:1547588. [PMID: 40196347 PMCID: PMC11973290 DOI: 10.3389/fmed.2025.1547588] [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: 12/18/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Background Early diagnosis of non-union fractures is vital for treatment planning, yet studies using bone morphometric parameters for this purpose are scarce. This study aims to create a fracture micro-CT image dataset, design a deep learning algorithm for fracture segmentation, and develop an early diagnosis model for fracture non-union. Methods Using fracture animal models, micro-CT images from 12 rats at various healing stages (days 1, 7, 14, 21, 28, and 35) were analyzed. Fracture lesion frames were annotated to create a high-resolution dataset. We proposed the Vision Mamba Triplet Attention and Edge Feature Decoupling Module UNet (VM-TE-UNet) for fracture area segmentation. And we extracted bone morphometric parameters to establish an early diagnostic evaluation system for the non-union of fractures. Results A dataset comprising 2,448 micro-CT images of the rat fracture lesions with fracture Region of Interest (ROI), bone callus and healing characteristics was established and used to train and test the proposed VM-TE-UNet which achieved a Dice Similarity Coefficient of 0.809, an improvement over the baseline's 0.765, and reduced the 95th Hausdorff Distance to 13.1. Through ablation studies, comparative experiments, and result analysis, the algorithm's effectiveness and superiority were validated. Significant differences (p < 0.05) were observed between the fracture and fracture non-union groups during the inflammatory and repair phases. Key indices, such as the average CT values of hematoma and cartilage tissues, BS/TS and BS/TV of mineralized cartilage, BS/TV of osteogenic tissue, and BV/TV of osteogenic tissue, align with clinical methods for diagnosing fracture non-union by assessing callus presence and local soft tissue swelling. On day 14, the early diagnosis model achieved an AUC of 0.995, demonstrating its ability to diagnose fracture non-union during the soft-callus phase. Conclusion This study proposed the VM-TE-UNet for fracture areas segmentation, extracted micro-CT indices, and established an early diagnostic model for fracture non-union. We believe that the prediction model can effectively screen out samples of poor fracture rehabilitation caused by blood supply limitations in rats 14 days after fracture, rather than the widely accepted 35 or 40 days. This provides important reference for the clinical prediction of fracture non-union and early intervention treatment.
Collapse
Affiliation(s)
- Hui Yu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University School of Medicine, Tianjin, China
| | - Qiyue Mu
- Department of Biomedical Engineering, Tianjin University School of Medicine, Tianjin, China
| | - Zhi Wang
- Radiology Department, Tianjin University Tianjin Hospital, Tianjin, China
| | - Yu Guo
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University School of Medicine, Tianjin, China
| | - Jing Zhao
- Department of Biomedical Engineering, Tianjin University School of Medicine, Tianjin, China
| | - Guangpu Wang
- Department of Biomedical Engineering, Tianjin University School of Medicine, Tianjin, China
| | - Qingsong Wang
- Department of Biomedical Engineering, Tianjin University School of Medicine, Tianjin, China
| | - Xianghong Meng
- Radiology Department, Tianjin University Tianjin Hospital, Tianjin, China
| | - Xiaoman Dong
- Radiology Department, Tianjin University Tianjin Hospital, Tianjin, China
| | - Shuo Wang
- Department of Biomedical Engineering, Tianjin University School of Medicine, Tianjin, China
| | - Jinglai Sun
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin University School of Medicine, Tianjin, China
| |
Collapse
|
9
|
Im GB, Lee JG, Lim H, Lee JW, Park HS, Kim Y, Asad N, Kim HR, Wie JJ, Bhang SH. Soft Pneumatic Device Designed to Mimic the Periosteal Environment for Regulating the Fate of Mesenchymal Stem Cells. Adv Healthc Mater 2025:e2403229. [PMID: 40123288 DOI: 10.1002/adhm.202403229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/16/2025] [Indexed: 03/25/2025]
Abstract
Replicating the complex mechanical forces of muscle movement and fluid flow in in vitro cell culture systems is crucial for understanding cell differentiation and development. However, previous research focused on cell differentiation on static micro/nanotextures without a force field or flat 2-dimensional substrates under a continuous in-plane mechanical force. In this study, cell differentiation is reported using a spatial geometric platform that can periodically modulate complex mechanical forces through a custom-made soft pneumatic device (SPD) to mimic the interfaces between periosteum and interstitial fluid. To elucidate fluidic dynamics and cell fates relevant to bone physiology, the platform exhibited distinct functional responses based on mechanical force levels: low mechanotransduction induced mesenchymal stem/progenitor cells differentiation into osteoprogenitor cells (≈1.5-fold increase in osteo-differentiation), while high mechanotransduction resulted in structural disruptions resembling cell detachment without protein degradation (≈2-fold increase in effective cell detachment). Numerical simulations of SPD elucidated the principal mechanical components for programmable cell differentiation and detachment by deconvoluting the in-plane and out-of-plane mechanical forces of the SPD complex mode. This study offers comprehensive and novel insights into the correlation between mechanical forces and cell differentiation, recovery, and injury in organisms.
Collapse
Affiliation(s)
- Gwang-Bum Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae Gyeong Lee
- Department of Organic and Nano Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Human-Tech Convergence Program, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hosub Lim
- Division of Engineering in Medicine and Renal Division, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Boston, MA, 02114, USA
| | - Jae-Won Lee
- School of Electronic and Electrical Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hyun Su Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yongju Kim
- Department of Polymer Science and Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - Nauman Asad
- School of Electronic and Electrical Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hak-Rin Kim
- School of Electronic and Electrical Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
- School of Electronics Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jeong Jae Wie
- Department of Organic and Nano Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Human-Tech Convergence Program, Hanyang University, Seoul, 04763, Republic of Korea
- The Research Institute of Industrial Science, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Chemical Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Chemical Engineering, State University of New York College of Environmental Science and Forestry, Syracuse, NY, 13210, USA
- The Michael M. Szwarc Polymer Research Institute, State University of New York College of Environmental Science and Forestry, Syracuse, NY, 13210, USA
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
10
|
Ellur G, Govindappa PK, Subrahmanian S, Romero GF, Gonzales DA, Margolis DS, Elfar JC. 4-Aminopyridine Promotes BMP2 Expression and Accelerates Tibial Fracture Healing in Mice. J Bone Joint Surg Am 2025:00004623-990000000-01397. [PMID: 40120116 DOI: 10.2106/jbjs.24.00311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
BACKGROUND Delayed bone healing is common in orthopaedic clinical care. Agents that alter cell function to enhance healing would change treatment paradigms. 4-aminopyridine (4-AP) is a U.S. Food and Drug Administration (FDA)-approved drug shown to improve walking in patients with chronic neurological disorders. We recently showed 4-AP's positive effects in the setting of nerve, wound, and even combined multi-tissue limb injury. Here, we directly investigated the effects of 4-AP on bone fracture healing, where differentiation of mesenchymal stem cells into osteoblasts is crucial. METHODS All animal experiments conformed to the protocols approved by the Institutional Animal Care and Use Committee at the University of Arizona and Pennsylvania State University. Ten-week-old C57BL/6J male mice (22 to 28 g), following midshaft tibial fracture, were assigned to 4-AP (1.6 mg/kg/day, intraperitoneal [IP]) and saline solution (0.1 mL/mouse/day, IP) treatment groups. Tibiae were harvested on day 21 for micro-computed tomography (CT), 3-point bending tests, and histomorphological analyses. 4-AP's effect on human bone marrow mesenchymal stem cell (hBMSC) and human osteoblast (hOB) cell viability, migration, and proliferation; collagen deposition; matrix mineralization; and bone-forming gene/protein expression analyses was assessed. RESULTS 4-AP significantly upregulated BMP2 gene and protein expression and gene expression of RUNX2, OSX, BSP, OCN, and OPN in hBMSCs and hOBs. 4-AP significantly enhanced osteoblast migration and proliferation, collagen deposition, and matrix mineralization. Radiographic and micro-CT imaging confirmed 4-AP's benefit versus saline solution treatment in mouse tibial fracture healing (bone mineral density, 687.12 versus 488.29 mg hydroxyapatite/cm3 [p ≤ 0.0021]; bone volume/tissue volume, 0.87 versus 0.72 [p ≤ 0.05]; trabecular number, 7.50 versus 5.78/mm [p ≤ 0.05]; and trabecular thickness, 0.08 versus 0.06 mm [p ≤ 0.05]). Three-point bending tests demonstrated 4-AP's improvement of tibial fracture biomechanical properties versus saline solution (stiffness, 27.93 versus 14.30 N/mm; p ≤ 0.05). 4-AP also increased endogenous BMP2 expression and matrix components in healing callus. CONCLUSIONS 4-AP increased the healing rate, biomechanical properties, and endogenous BMP2 expression of tibiae following fracture. LEVEL OF EVIDENCE Prognostic Level III. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Govindaraj Ellur
- Department of Orthopaedics and Sports Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Prem Kumar Govindappa
- Department of Orthopaedics and Sports Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | | | - Gerardo Figueroa Romero
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Arizona College of Medicine, Tucson, Arizona
| | - David A Gonzales
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Arizona College of Medicine, Tucson, Arizona
| | - David S Margolis
- Department of Orthopaedics and Sports Medicine, University of Arizona College of Medicine, Tucson, Arizona
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Arizona College of Medicine, Tucson, Arizona
| | - John C Elfar
- Department of Orthopaedics and Sports Medicine, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
11
|
Hoveidaei AH, Sadat-Shojai M, Nabavizadeh SS, Niakan R, Shirinezhad A, MosalamiAghili S, Tabaie S. Clinical challenges in bone tissue engineering - A narrative review. Bone 2025; 192:117363. [PMID: 39638083 DOI: 10.1016/j.bone.2024.117363] [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: 09/15/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Bone tissue engineering (BTE) has emerged as a promising approach to address large bone defects caused by trauma, infections, congenital malformations, and tumors. This review focuses on scaffold design, cell sources, growth factors, and vascularization strategies, highlighting their roles in developing effective treatments. We explore the complexities of balancing mechanical properties, porosity, and biocompatibility in scaffold materials, alongside optimizing mesenchymal stem cell delivery methods. The critical role of growth factors in bone regeneration and the need for controlled release systems are discussed. Vascularization remains a significant hurdle, with strategies such as angiogenic factors, co-culture systems, and bioprinting under investigation. Mechanical challenges, tissue responses, and inflammation management are examined, alongside gene therapy's potential for enhancing osteogenesis and angiogenesis via both viral and non-viral delivery methods. The review emphasizes the impact of patient-specific factors on bone healing outcomes and the importance of personalized approaches. Future directions are described, emphasizing the necessity of interdisciplinary cooperation to advance the field of BTE and convert laboratory results into clinically feasible solutions.
Collapse
Affiliation(s)
- Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA.
| | - Mehdi Sadat-Shojai
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran.
| | - Sara S Nabavizadeh
- Otolaryngology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Niakan
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Sean Tabaie
- Department of Orthopaedic Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
12
|
Cao Y, Liu C, Ye W, Zhao T, Fu F. Functional Hydrogel Interfaces for Cartilage and Bone Regeneration. Adv Healthc Mater 2025; 14:e2403079. [PMID: 39791312 DOI: 10.1002/adhm.202403079] [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: 08/16/2024] [Revised: 11/08/2024] [Indexed: 01/12/2025]
Abstract
Effective treatment of bone diseases is quite tricky due to the unique nature of bone tissue and the complexity of the bone repair process. In combination with biological materials, cells and biological factors can provide a highly effective and safe treatment strategy for bone repair and regeneration, especially based on these multifunctional hydrogel interface materials. However, itis still a challenge to formulate hydrogel materials with fascinating properties (e.g., biological activity, controllable biodegradability, mechanical strength, excellent cell/tissue adhesion, and controllable release properties) for their clinical applications in complex bone repair processes. In this review, we will highlight recent advances in developing functional interface hydrogels. We then discuss the barriers to producing of functional hydrogel materials without sacrificing their inherent properties, and potential applications in cartilage and bone repair are discussed. Multifunctional hydrogel interface materials can serve as a fundamental building block for bone tissue engineering.
Collapse
Affiliation(s)
- Yucheng Cao
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Changyi Liu
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Wenjun Ye
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Tianrui Zhao
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Fanfan Fu
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| |
Collapse
|
13
|
Sembadani YM, Barus L. Biomechanical perspective of dynamic elastic therapy and intermaxillary fixation in concomitant and single site mandibular fracture: Serial case report. Int J Surg Case Rep 2025; 128:110912. [PMID: 39908931 PMCID: PMC11847546 DOI: 10.1016/j.ijscr.2025.110912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
INTRODUCTION AND IMPORTANCE Mandibular fractures are one of the most common sites of injuries and are difficult to restore in establishing preinjury occlusion and it is influenced by biomechanical principles. Open Reduction and Internal Fixation remain the gold standard in treating mandibular fractures to achieve pre-injury occlusion though it was limited toward expensive cost. As an option, closed reduction using an arch bar, wire, and elastic combination was preferred to establish functional preinjury occlusion. CASES PRESENTATION Patients came to University Hospital with chief complaints of pain and difficulties in mouth opening and mastication. From clinical and radiographic examination the first case with history of assault injury during martial arts was found unilateral left mandibular angle and right mandibular body fracture. In the second case with the history of motorcycle injury 11 days ago was found displaced right parasimphysis mandibular fracture. CLINICAL DISCUSSION All cases was treated with close reduction using dynamic elastic for gradual reposition and followed with wire fixation until occlusion was established. After the evaluation, functional occlusion and normal mouth opening was achieved. CONCLUSION The selection of the therapy on multiple site fractures can be handled in a closed reduction with Intermaxillary Fixation which minimally invasive, low risk of nerve injury and give a good bone remodeling process and can restore mandibular function according to anatomy.
Collapse
Affiliation(s)
| | - Liska Barus
- Department of Oral And Maxillofacial Surgery, Universitas Airlangga, Surabaya, Indonesia.
| |
Collapse
|
14
|
Wu X, Yang H, Liu G, Sun W, Li J, Zhao Y, Gao X, Liu X, Song F, Wang S, Lu Z, Chen C, Huang C. Osteomimix: A Multidimensional Biomimetic Cascade Strategy for Bone Defect Repair. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416715. [PMID: 39924776 DOI: 10.1002/adma.202416715] [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: 10/31/2024] [Revised: 01/24/2025] [Indexed: 02/11/2025]
Abstract
Despite advancements in biomimetic mineralization techniques, the repair of large-scale bone defects remains a significant challenge. Inspired by the bone formation process, a multidimensional biomimetic cascade strategy is developed by replicating the biomineralization cascade, emulating the hierarchical structure of bone, and biomimicking its biological functions for efficient bone regeneration. This strategy involves the photocrosslinking of sodium methacrylate carboxymethyl cellulose-stabilized amorphous magnesium-calcium phosphate with methacrylate-modified type I collagen to create a self-mineralizing hydrogel. The hydrogel is then integrated with either naturally derived or synthetic oriented bulk scaffolds. The resulting composite, named Osteomimix, provides excellent mechanical support and can be customized for irregular bone defects using CAD/CAM technology. Through in vitro and in vivo studies, this work finds that Osteomimix exhibits spontaneous in situ biomimetic mineralization in a cell-free environment, while modulating immune responses and promoting vascularized bone formation in a cell-dependent manner. Built on bone-specific insights, this strategy achieves biomimicry across temporal, spatial, and functional dimensions, facilitating the seamless integration of artificial constructs with the natural tissue repair dynamics.
Collapse
Affiliation(s)
- Xiaoyi Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, School of Resource and Environmental Sciences, Wuhan University, Wuhan, 430079, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Gufeng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Wei Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jiyun Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yaning Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xin Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xuzheng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Shilei Wang
- Hubei Engineering Center of Natural Polymer-Based Medical Materials and Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Ziyang Lu
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, School of Resource and Environmental Sciences, Wuhan University, Wuhan, 430079, China
| | - Chaoji Chen
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, School of Resource and Environmental Sciences, Wuhan University, Wuhan, 430079, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
15
|
Brazeau D, Deshaies AA, Williamson D, Bernard F, Arbour C, Pinard AM, Rouleau D, De Beaumont L. Impact of an acute 1-month cannabidiol treatment on pain and inflammation after a long bone fracture: a triple-blind randomised, placebo-controlled, clinical trial protocol. BMJ Open 2025; 15:e092919. [PMID: 39979051 PMCID: PMC11842986 DOI: 10.1136/bmjopen-2024-092919] [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: 08/26/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
INTRODUCTION Acute pain levels following orthopaedic injury (eg, fracture) are a predictor of the onset of chronic pain, which affects nearly 50% of fracture patients and impairs functional recovery. Among current pharmacological treatments for acute pain, non-steroidal anti-inflammatory drugs have been associated with delayed bone healing, while opioids inhibit effective bone remodelling, increase the risk of pseudarthrosis and carry a high risk of addiction. In light of this, the development of new pain treatments is essential. Cannabidiol (CBD), a non-addictive and non-psychotropic cannabis component stands out as a potential therapeutic agent, given its analgesic and anti-inflammatory properties as well as its potential benefits for bone healing. This randomised controlled trial aims to investigate the effect of acute CBD treatment, compared with placebo, on patients' self-reported pain, inflammation and well-being following a fracture injury. METHODS AND ANALYSIS This is a triple-blind, randomised, placebo-controlled clinical trial. A total of 225 adults aged 18-70 years, who have suffered a long bone fracture and were treated at the Hôpital du Sacré-Coeur de Montréal, will be randomly assigned within 1 week to one of three treatment arms (25 mg or 50 mg of CBD or placebo) for 1 month. The primary outcome will be the difference in the pain score between groups at 1-month follow-up. Secondary outcomes will include measures of persistent pain, inflammation, opioid usage, quality of life, sleep quality, depression, anxiety, cognition and orthopaedic function. Data will be collected at baseline, 1-month and 3-month follow-ups. ETHICS AND DISSEMINATION This study obtained a Health Canada licence for use of cannabis products. It has also been approved by Health Canada and the Research Ethics Board of the CIUSSS du Nord-de-l'Île-de-Montréal (Project ID 2025-2105). The findings will be published in a peer-reviewed journal and presented at local, national and international conferences. The trial's results will be made publicly available on the ClinicalTrials.gov database. TRIAL REGISTRATION NUMBER NCT06448923.
Collapse
Affiliation(s)
- Daphnée Brazeau
- Psychology, University of Montreal, Montreal, Quebec, Canada
- CIUSSS du Nord-de-l'Ile-de-Montreal, Montreal, Quebec, Canada
| | | | - David Williamson
- CIUSSS du Nord-de-l'Ile-de-Montreal, Montreal, Quebec, Canada
- Pharmacie, University of Montreal, Montreal, Quebec, Canada
| | - Francis Bernard
- Hopital du Sacre-Coeur de Montreal, Montreal, Quebec, Canada
- Medecine Faculty, University of Montreal, Montreal, Quebec, Canada
| | - Caroline Arbour
- CIUSSS du Nord-de-l'Ile-de-Montreal, Montreal, Quebec, Canada
- Nursing, University of Montreal, Montreal, Quebec, Canada
| | | | | | - Louis De Beaumont
- CIUSSS du Nord-de-l'Ile-de-Montreal, Montreal, Quebec, Canada
- Department of Surgery, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Wu S, Lu J. Liposome-Enabled Nanomaterials for Muscle Regeneration. SMALL METHODS 2025:e2402154. [PMID: 39967365 DOI: 10.1002/smtd.202402154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/08/2025] [Indexed: 02/20/2025]
Abstract
Muscle regeneration is a vital biological process that is crucial for maintaining muscle function and integrity, particularly for the treatment of muscle diseases such as sarcopenia and muscular dystrophy. Generally, muscular tissues can self-repair and regenerate under various conditions, including acute or chronic injuries, aging, and genetic mutation. However, regeneration becomes challenging beyond a certain threshold, particularly in severe muscle injuries or progressive diseases. In recent years, liposome-based nanotechnologies have shown potential as promising therapeutic strategies for muscle regeneration. Liposomes offer an adaptable platform for targeted drug delivery due to their cell membrane-like structure and excellent biocompatibility. They can enhance drug solubility, stability, and targeted delivery while minimizing systemic side effects by different mechanisms. This review summarizes recent advancements, discusses current applications and mechanisms, and highlights challenges and future directions for possible clinical translation of liposome-based nanomaterials in the treatment of muscle diseases. It is hoped this review offers new insights into the development of liposome-enabled nanomedicine to address current limitations.
Collapse
Affiliation(s)
- Shuang Wu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
- Clinical and Translational Oncology Program, The University of Arizona Cancer Center, Tucson, AZ, 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
17
|
Liu F, Yang F, Guo L, Yang M, Li Y, Li J, Guo Y, He S. Alteration in Tracheal Morphology and Transcriptomic Features in Calves After Infection with Mycoplasma bovis. Microorganisms 2025; 13:442. [PMID: 40005807 PMCID: PMC11857948 DOI: 10.3390/microorganisms13020442] [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: 01/21/2025] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Mycoplasma bovis is one of the most important pathogens in animal husbandry, and the current infection and morbidity rates are increasing year by year, causing great losses to the farming industry and seriously affecting animal welfare. In this study, we took tracheal tissues from calves infected with M. bovis to make pathological tissue sections for observation, and selected tracheal tissues for transcriptome sequencing to screen differentially expressed genes based on the threshold |log2FoldChange| > 1 and Padjust < 0.05 and functional enrichment, to explore in depth the potential mechanisms of bovine tracheal damage caused by bovine tracheitis. Experiments were conducted to observe the changes in tracheal tissues after M. bovis infection through pathological sections of the trachea of M. bovis-infected calves. From the transcriptome sequencing results, we mined the main differential genes and important metabolic pathways of M. bovis causing damage to the trachea of calves. It was found that the cricoid cartilage tissue of the trachea was congested and hemorrhagic after M. bovis infection in calves, and the pathological sections showed localized necrosis of epithelial cells, disorganization, high inflammatory cell infiltration in the interepithelial and lamina propria, and some epithelial cell detachment. Transcriptome sequencing identified 4199 DEGs, including 1378 up-regulated genes and 2821 down-regulated genes. KEGG enrichment analysis indicated that the differential genes were enriched to 59 significantly differing signaling pathways, and a number of important metabolic pathways related to tracheitis induced by M. bovis-infected calves were unearthed. The major ones included IL-17, the Toll-like receptor, JAK/STAT, the PI3K-Akt signaling pathway, etc. In this study, we found that M. bovis infection of calves caused inflammatory damage to the trachea, and transcriptome sequencing results also showed significant differences in the expression of key genes such as IL-6 inflammatory factor, CASP8, and APOA1.
Collapse
Affiliation(s)
- Fan Liu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (F.L.); (F.Y.); (L.G.); (M.Y.); (J.L.)
- Institute of Animal Sciences, Ningxia Academy of Agricultural and Forestry Sciences, Yinchuan 750002, China
| | - Fei Yang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (F.L.); (F.Y.); (L.G.); (M.Y.); (J.L.)
| | - Lei Guo
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (F.L.); (F.Y.); (L.G.); (M.Y.); (J.L.)
| | - Mengmeng Yang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (F.L.); (F.Y.); (L.G.); (M.Y.); (J.L.)
| | - Yong Li
- College of Life Science and Technology, Ningxia Polytechnic, Yinchuan 750002, China;
| | - Jidong Li
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (F.L.); (F.Y.); (L.G.); (M.Y.); (J.L.)
| | - Yanan Guo
- Institute of Animal Sciences, Ningxia Academy of Agricultural and Forestry Sciences, Yinchuan 750002, China
| | - Shenghu He
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (F.L.); (F.Y.); (L.G.); (M.Y.); (J.L.)
| |
Collapse
|
18
|
Ghanem W, Ezzeddine H, Saad R, Kiwan E, Dahdouh R, Fakih O, Sakhat G, Alam E, Najjar J, Assaf F, Chahine M, Dib N, Kortbawi R, Badra M, Moucharafieh R. State of the Nonunion: A review of the latest literature. Orthop Rev (Pavia) 2025; 17:129085. [PMID: 39925644 PMCID: PMC11807701 DOI: 10.52965/001c.129085] [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: 10/29/2024] [Accepted: 12/06/2024] [Indexed: 02/11/2025] Open
Abstract
The distinction between prolonged bone healing and nonunion in long bone fracture remains a historical challenge in the field of orthopedics. Despite numerous proposed definitions and scoring systems, a consensus remains elusive, thereby complicating both diagnosis and treatment. An accurate diagnosis is necessary, facilitated by a range of imaging modalities. Bone nonunion management encompasses surgical and non-surgical options, including external or internal fixation, and bone grafting, tailored to the nonunion type. This review discusses the pathophysiology of nonunion, risk factors, diagnosis and treatment. It particularly addresses early detection and the impacts of nonunion on the patient. The aim of this review is to obtain a global and updated point of view regarding nonunion of the bone as well as to reflect on the potential use of untraditional methods in their treatment such as orthobiologics, along with emerging and non-invasive technologies including shockwave therapy, gene therapy, tissue engineering, regenerative medicine and 3D printing.
Collapse
Affiliation(s)
- Wendy Ghanem
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Hady Ezzeddine
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Rita Saad
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Elyssa Kiwan
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Raymonde Dahdouh
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Omar Fakih
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Georges Sakhat
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Elie Alam
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Joseph Najjar
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Fouad Assaf
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Moro Chahine
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Nabil Dib
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Rabih Kortbawi
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
| | - Mohammad Badra
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
- Department of Orthopedics and Traumatology, Clemenceau Medical, Beirut, Lebanon
| | - Ramzi Moucharafieh
- Department of Orthopedics and Traumatology, Faculty of Medicine University of Balamand
- Department of Orthopedics and Traumatology, Clemenceau Medical, Beirut, Lebanon
| |
Collapse
|
19
|
Perepletchikova D, Malashicheva A. Communication between endothelial cells and osteoblasts in regulation of bone homeostasis: Notch players. Stem Cell Res Ther 2025; 16:56. [PMID: 39920854 PMCID: PMC11806792 DOI: 10.1186/s13287-025-04176-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: 07/25/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
Endothelial cells coat blood vessels and release molecular signals to affect the fate of other cells. Endothelial cells can adjust their behavior in response to the changing microenvironmental conditions. During bone regeneration, bone tissue cells release factors that promote blood vessel growth. Notch is a key signaling that regulates cell fate decisions in many tissues and plays an important role in bone tissue development and homeostasis. Understanding the interplay between angiogenesis and osteogenesis is currently a focus of research efforts in order to facilitate and improve osteogenesis when needed. Our review explores the cellular and molecular mechanisms including Notch-dependent endothelial-MSC communication that drive osteogenesis-angiogenesis processes and their effects on bone remodeling and repair.
Collapse
Affiliation(s)
| | - Anna Malashicheva
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia, 194064.
| |
Collapse
|
20
|
Fan H, Xue B, Lu J, Sun T, Zhao Q, Liu Y, Niu M, Yu S, Yang Y, Zhang L. Recent advances of bioaerogels in medicine: Preparation, property and application. Int J Biol Macromol 2025; 291:139144. [PMID: 39722377 DOI: 10.1016/j.ijbiomac.2024.139144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/14/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Bioaerogels represent a type of three-dimensional porous materials fabricated from natural biopolymers, and show a significant potential for medical application due to their characteristics of extremely low density, high specific surface area, excellent biocompatibility and biodegradability. The preparation method and parameters of bioaerogels are focused on, and their influence on the structure and properties of bioaerogels are discussed in detail. Then, to match the properties of bioaerogels with the medical applications, this work emphasizes the main properties (including biocompatibility, degradability, and mechanical properties), structural parameters (such as suitable porosity, pore size and high specific surface area), and further summarizes the influence of single-component and composite bioaerogels on their properties. Moreover, according to the different applications (wound healing, drug delivery, and tissue engineering and other fields), the function method, mechanism and practical effect of bioaerogels are comprehensively analyzed. Finally, the challenges, future research directions, and solutions for the practical application of bioaerogels in medicine are discussed.
Collapse
Affiliation(s)
- Haoyong Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Baoxia Xue
- College of Textile Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jiaxin Lu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Tao Sun
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Qinke Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Yong Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Mei Niu
- College of Textile Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Shiping Yu
- Department of Interventional Therapy, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030001, China
| | - Yongzhen Yang
- Key Laboratory of Interface Science and Engineering in Advanced Materials, Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China.
| | - Li Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China.
| |
Collapse
|
21
|
Wang Q, Gao Y, Chen Y, Wang X, Pei Q, Zhang T, Wang C, Pan J. Synergistic Enhancement of Antibacterial and Osteo-Immunomodulatory Activities of Titanium Implants via Dual-Responsive Multifunctional Surfaces. Adv Healthc Mater 2025; 14:e2404260. [PMID: 39690750 DOI: 10.1002/adhm.202404260] [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: 10/31/2024] [Revised: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Bone implant-associated infections and inflammations, primarily caused by bacteria colonization, frequently result in unsuccessful procedures and pose significant health risks to patients. To mitigate these challenges, the development of engineered implants with spatiotemporal regulation capabilities, designed to inhibit bacterial survival and modulate immune responses in the early stage, while promoting bone defect healing in the late stage is proposed. The implants are functionalized with ε-poly-l-lysine-phenylboronic acid (PP) via dynamic boronic ester bonds, which facilitate its release through a reactive oxygen species (ROS) and pH-responsive strategy, thereby establishing an antibacterial microenvironment on and around the implants. Additionally, the dynamic metal coordination interaction facilitates the loading and sustained release of Sr2+ under an acidic environment, providing immunomodulatory and osteogenic effects. The ROS/pH-responsive feature, coupled with the implant-bone tissue integration process, affords precise spatiotemporal regulation of the Ti-TA-Sr-PP implants. This strategy represents a promising approach for the preparation of advanced bone implants.
Collapse
Affiliation(s)
- Qing Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ya Gao
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yanzheng Chen
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Xuan Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qingguo Pei
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Taiyu Zhang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Changping Wang
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jinsong Pan
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
22
|
Liu L, Chen H, Zhao X, Han Q, Xu Y, Liu Y, Zhang A, Li Y, Zhang W, Chen B, Wang J. Advances in the application and research of biomaterials in promoting bone repair and regeneration through immune modulation. Mater Today Bio 2025; 30:101410. [PMID: 39811613 PMCID: PMC11731593 DOI: 10.1016/j.mtbio.2024.101410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/02/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
With the ongoing development of osteoimmunology, increasing evidence indicates that the local immune microenvironment plays a critical role in various stages of bone formation. Consequently, modulating the immune inflammatory response triggered by biomaterials to foster a more favorable immune microenvironment for bone regeneration has emerged as a novel strategy in bone tissue engineering. This review first examines the roles of various immune cells in bone tissue injury and repair. Then, the contributions of different biomaterials, including metals, bioceramics, and polymers, in promoting osteogenesis through immune regulation, as well as their future development directions, are discussed. Finally, various design strategies, such as modifying the physicochemical properties of biomaterials and integrating bioactive substances, to optimize material design and create an immune environment conducive to bone formation, are explored. In summary, this review comprehensively covers strategies and approaches for promoting bone tissue regeneration through immune modulation. It offers a thorough understanding of current research trends in biomaterial-based immune regulation, serving as a theoretical reference for the further development and clinical application of biomaterials in bone tissue engineering.
Collapse
Affiliation(s)
- Li Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Hao Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Xue Zhao
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Qing Han
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Yongjun Xu
- Department of Orthopedics Surgery, Wangqing County People's Hospital, Yanbian, 133000, Jilin, China
| | - Yang Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Aobo Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Yongyue Li
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Weilong Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Bingpeng Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Jincheng Wang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| |
Collapse
|
23
|
Zhang Y, Zhou C, Xie Q, Xia L, Liu L, Bao W, Lin H, Xiong X, Zhang H, Zheng Z, Zhao J, Liang W. Dual release scaffolds as a promising strategy for enhancing bone regeneration: an updated review. Nanomedicine (Lond) 2025; 20:371-388. [PMID: 39891431 PMCID: PMC11812394 DOI: 10.1080/17435889.2025.2457317] [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: 01/20/2025] [Indexed: 02/03/2025] Open
Abstract
Advancements in tissue regeneration, particularly bone regeneration is key area of research due to potential of novel therapeutic approaches. Efforts to reduce reliance on autologous and allogeneic bone grafts have led to the development of biomaterials that promote synchronized and controlled bone healing. However, the use of growth factors is limited by their short half-life, slow tissue penetration, large molecular size and potential toxicity. These factors suggest that traditional delivery methods may be inadequate hence, to address these challenges, new strategies are being explored. These novel approaches include the use of bioactive substances within advanced delivery systems that enable precise spatiotemporal control. Dual-release composite scaffolds offer a promising solution by reducing the need for multiple surgical interventions and simplifying the treatment process. These scaffolds allow for sustained and controlled drug release, enhancing bone repair while minimizing the drawbacks of conventional methods. This review explores various dual-drug release systems, discussing their modes of action, types of drugs used and release mechanisms to improve bone regeneration.
Collapse
Affiliation(s)
- Yongtao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, Zhejiang, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
24
|
Jeong H, Subramanian K, Lee JB, Byun H, Shin H, Yun JH. Anti-inflammatory and osteoconductive multi-functional nanoparticles for the regeneration of an inflamed alveolar bone defect. Biomater Sci 2025; 13:810-825. [PMID: 39749408 DOI: 10.1039/d4bm01280a] [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: 01/04/2025]
Abstract
Infected alveolar bone defects pose challenging clinical issues due to disrupted intrinsic healing mechanisms. Thus, the employment of advanced biomaterials enabling the modulation of several aspects of bone regeneration is necessary. This study investigated the effect of multi-functional nanoparticles on anti-inflammatory/osteoconductive characteristics and bone repair in the context of inflamed bone abnormalities. Tannic-acid mineral nanoparticles (TMPs) were prepared by the supramolecular assembly of tannic acid with bioactive calcium and phosphate ions, which were subsequently incorporated into collagen plugs via molecular interactions. Under physiological conditions, in vitro analysis confirmed that tannic acid was dissociated and released, which significantly reduced the expression of pro-inflammatory genes in lipopolysaccharide (LPS)-activated RAW264.7 cells. Meanwhile, the bioactive ions of Ca2+ and PO43- synergistically increased the gene and protein expressions of osteogenic markers of bone marrow-derived stem cells. For in vivo studies, combined endodontic-periodontal lesions were induced in beagle dogs where the plugs were readily implanted. After 2 months of the implantation, analysis of micro-computed tomography and histomorphometry revealed that groups of dogs implanted with the plug incorporating TMPs exhibited a significant decrease in bone surface density as well as structural model index, and significant increase in the mineralized bone content, respectively, with positive OPN expression being observed in reversal lines. Notably, the profound improvement in bone regeneration relied on the concentration of TMPs in the implants, underscoring the promise of multi-functional nanoparticles for treating infected alveolar bones.
Collapse
Affiliation(s)
- Hyewoo Jeong
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea.
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Keerthana Subramanian
- Department of Periodontology, College of Dentistry and Institute of Oral Bioscience, Jeonbuk National University, Jeonju, Republic of Korea.
| | - Jong-Bin Lee
- Department of Periodontology, College of Dentistry and Research Institute of Oral Sciences, Gangneung-Wonju National University, Gangneung, Republic of Korea
| | - Hayeon Byun
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea.
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea.
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul, Republic of Korea
| | - Jeong-Ho Yun
- Department of Periodontology, College of Dentistry and Institute of Oral Bioscience, Jeonbuk National University, Jeonju, Republic of Korea.
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| |
Collapse
|
25
|
Gallo MC, Elias A, Reynolds J, Ball JR, Lieberman JR. Regional Gene Therapy for Bone Tissue Engineering: A Current Concepts Review. Bioengineering (Basel) 2025; 12:120. [PMID: 40001640 PMCID: PMC11852166 DOI: 10.3390/bioengineering12020120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
The management of segmental bone defects presents a complex reconstruction challenge for orthopedic surgeons. Current treatment options are limited by efficacy across the spectrum of injury, morbidity, and cost. Regional gene therapy is a promising tissue engineering strategy for bone repair, as it allows for local implantation of nucleic acids or genetically modified cells to direct specific protein expression. In cell-based gene therapy approaches, a variety of different cell types have been described including mesenchymal stem cells (MSCs) derived from multiple sources-bone marrow, adipose, skeletal muscle, and umbilical cord tissue, among others. MSCs, in particular, have been well studied, as they serve as a source of osteoprogenitor cells in addition to providing a vehicle for transgene delivery. Furthermore, MSCs possess immunomodulatory properties, which may support the development of an allogeneic "off-the-shelf" gene therapy product. Identifying an optimal cell type is paramount to the successful clinical translation of cell-based gene therapy approaches. Here, we review current strategies for the management of segmental bone loss in orthopedic surgery, including bone grafting, bone graft substitutes, and operative techniques. We also highlight regional gene therapy as a tissue engineering strategy for bone repair, with a focus on cell types and cell sources suitable for this application.
Collapse
Affiliation(s)
- Matthew C. Gallo
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Aura Elias
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Julius Reynolds
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Jacob R. Ball
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Jay R. Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
26
|
Sun B, Thangavelu V, Yakubov R, Sun C, Khan M, Chaudhari S, Sheth U. The impact of preexisting psychiatric disorders on patient outcomes following primary total shoulder arthroplasty: A systematic review and quantitative synthesis. Shoulder Elbow 2025:17585732251314130. [PMID: 39866904 PMCID: PMC11758438 DOI: 10.1177/17585732251314130] [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: 09/19/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025]
Abstract
Purpose To summarize complication rates, reoperation rates, length-of-stay (LOS), patient-reported outcome measures (PROMs), and range of motion following total shoulder arthroplasty (TSA) in patients with preexisting psychiatric disorders (PDs) compared to controls. Methods Three databases (MEDLINE, PubMed, and EMBASE) were searched from inception to 4 March 2024 to identify studies comparing outcomes between patients undergoing anatomic (aTSA) or reverse TSA (rTSA) with or without a preexisting psychiatric condition. The authors adhered to the preferred reporting items for systematic reviews and meta-analyses and revised assessment of multiple systematic review guidelines. Data on demographics, as well as postoperative complication rates, reoperation rates, LOS, PROMs, and range of motion were extracted from included studies. PROMs included the American Shoulder and Elbow Surgeons (ASESs) score, and visual analogue scale (VAS) pain score. Meta-analyses were conducted for outcomes reported by multiple studies, with odds ratios (ORs) and mean differences (MDs) as effect measures for continuous and dichotomous outcomes, respectively. Results Thirteen studies were included in this review, comprising a total of 820,831 TSA patients. The PD group (71.0% female) consisted of 150,432 patients (mean age: 67.6 ± 9.9) with a mean follow-up time of 34.1 ± 30.1 months. The control group (58.1% female) consisted of 670,399 patients (mean age: 69.4 ± 10.7) with a mean follow-up time of 39.1 ± 36.0 months. The PD group had significantly higher rates of complications and reoperation. The PD group also reported significantly lower postoperative ASES scores, higher postoperative VAS scores, and inferior postoperative abduction. There were no significant differences in postoperative LOS, forward flexion, internal rotation, or external rotation. Conclusion Patients with preexisting PDs may have a one-and-a-half times higher odds of postoperative complication or reoperation, as well as significantly worse postoperative pain and PROMs. Identification of at-risk individuals with preexisting psychiatric conditions and preoperative referral to a mental health specialist to optimize psychiatric conditions may benefit this patient cohort ahead of their shoulder arthroplasty procedure. Level of evidence IV.
Collapse
Affiliation(s)
- Bryan Sun
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - Vetri Thangavelu
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Rose Yakubov
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Clare Sun
- Faculty of Health Sciences, Queen’s University, Kingston, ON, Canada
| | - Moin Khan
- Division of Orthopaedic Surgery, Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Sumit Chaudhari
- Department of Psychiatry, London Health Sciences Centre, Western University, London, ON, Canada
| | - Ujash Sheth
- Sunnybrook Orthopaedic Upper Limb (SOUL), Sunnybrook Health Sciences Centre, Division of Orthopaedic Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Marom H, Khan MA, Darvish N, Tornetta III P, Khoury A, Weil YA, Skelley NWM, Allison DC, Meiron S, Ehrmann Barr T. β-Caryophyllene and Statins in Bone Fracture Healing - A Narrative Review. Orthop Res Rev 2025; 17:31-42. [PMID: 39872403 PMCID: PMC11771162 DOI: 10.2147/orr.s506427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025] Open
Abstract
Bone fractures are a leading cause of morbidity and healthcare expenditure globally. The complex healing process involves inflammation, cartilage formation, mineralization, and bone remodeling. Current treatments like immobilization, surgery, and bone grafting, though effective, pose significant challenges, such as prolonged recovery and high costs. Emerging therapies such as biological agents, pharmacological treatments, and physical stimulation techniques are also associated with high costs, side effects, and practical applicability limitations. There is a critical need for alternative therapies that are cost-effective, safe, and easy to use. Recent studies suggest the potential of β-caryophyllene (BCP) and statins in promoting bone healing. BCP, a naturally occurring anti-inflammatory and antioxidant compound found in essential oils, enhances osteoblast activity and inhibits osteoclastogenesis. Statins, known for their cholesterol-lowering effects, also promote bone formation and reduce bone resorption through multiple biochemical pathways. Both BCP and statins have shown promising results in preclinical studies, enhancing bone density and promoting fracture healing. This review explores the individual and potential synergistic effects of BCP and statins on bone fracture healing. It highlights the complementary mechanisms of these agents: BCP's anti-inflammatory and osteogenic properties and statins' ability to inhibit osteoclast activity and promote angiogenesis. Combining BCP and statins could offer a multifaceted approach to enhance fracture healing, reduce complications, and improve patient outcomes. While individual effects are supported preclinically, further studies investigating synergies, formulations, and clinical translation are needed to develop this promising novel therapeutic approach for improving fracture repair outcomes.
Collapse
Affiliation(s)
| | | | - Nissim Darvish
- Corporate Office, OrthoTreat Ltd, Tel Aviv-Jaffa, Israel
| | - Paul Tornetta III
- Department of Orthopaedic Surgery and Orthopaedic Trauma, Chobanian and Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Amal Khoury
- Division of Orthopaedic Surgery, Orthopaedic, and Reconstructive Trauma Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yoram A Weil
- Department of Orthopaedics, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | | | - Daniel C Allison
- Department of Orthopaedic Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sahar Meiron
- Corporate Office, OrthoTreat Ltd, Tel Aviv-Jaffa, Israel
| | | |
Collapse
|
28
|
Fonseca AC, Colavite PM, Azevedo MDCS, Passadori DC, Melchiades JL, Ortiz RC, Rodini CO, Trombone APF, Garlet GP. Inhibition of MEK1/2 Signaling Pathway Limits M2 Macrophage Polarization and Interferes in the Dental Socket Repair Process in Mice. BIOLOGY 2025; 14:107. [PMID: 40001875 PMCID: PMC11851886 DOI: 10.3390/biology14020107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/27/2024] [Accepted: 01/16/2025] [Indexed: 02/27/2025]
Abstract
Dental socket repair theoretically involves a constructive inflammatory immune response, which evolves from an initial M1 prevalence to a subsequent M2 dominance. In this scenario, the MEK1/2 signaling pathway is allegedly involved in M2 polarization. This study aimed to evaluate the impact of MEK1/2 pharmacological inhibition in the local host response and repair outcome. C57Bl/6-WT 8-week-old male mice were submitted to the extraction of the right upper incisor and treated (or not, control group) with MEK1/2 inhibitor PD0325901 (10 mg/kg/24 h/IP, MEK1/2i group) and analyzed at 0, 3, 7, and 14 days using microcomputed tomography, histomorphometry, birefringence, immunohistochemistry, and PCR array analysis. The results demonstrate that MEK1/2 inhibition limits the development of M2 response over time, being associated with lower expression of M2, MSCs, and bone markers, lower levels of growth and osteogenic factors, along with a higher expression of iNOS, IL-1b, IL-6, and TNF-α, as well inflammatory chemokines, indicating a predominantly M1 pro-inflammatory environment. This modulation of local inflammatory immune response is associated with impaired bone formation as demonstrated by microtomographic and histomorphometric data. The results show that MEK1/2 inhibition delays bone repair after tooth extraction, supporting the concept that M2 macrophages are essential elements for host response regulation and proper repair.
Collapse
Affiliation(s)
- Angélica Cristina Fonseca
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Priscila Maria Colavite
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Michelle de Campos Soriani Azevedo
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Daniela Carignatto Passadori
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Jessica Lima Melchiades
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Rafael Carneiro Ortiz
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Camila Oliveira Rodini
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Ana Paula Favaro Trombone
- Department of Health Sciences, Centro Universitário Sagrado Coração—UNISAGRADO, Bauru 17011-160, SP, Brazil;
| | - Gustavo Pompermaier Garlet
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| |
Collapse
|
29
|
Wang X, Zhang X, Gong C, Yang J, Chen J, Guo W. Functionalized GelMA/CMCS Composite Hydrogel Incorporating Magnesium Phosphate Cement for Bone Regeneration. Biomedicines 2025; 13:257. [PMID: 40002671 PMCID: PMC11852312 DOI: 10.3390/biomedicines13020257] [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: 12/11/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Bone regeneration remains a challenging issue in tissue engineering. The use of hydrogels as scaffolds for bone tissue repair has gained attention due to their biocompatibility and ability to mimic the extracellular matrix. This study aims to develop a functionalized GelMA/CMCS composite hydrogel incorporating magnesium phosphate cement (MPC) for enhanced bone regeneration. Methods: These composites were developed by incorporating potassium magnesium phosphate hexahydrate (KMgPO4·6H2O, MPC) powders into methacrylated gelatin/carboxymethyl chitosan (GelMA-C) hydrogels. The material's mechanical properties, antibacterial performance, and cytocompatibility were evaluated. In vitro experiments involved cell viability and osteogenic differentiation assays using rBMSCs as well as angiogenic potential assays using HUVECs. The hydrogel was also assessed for its potential in promoting bone repair in a rat (Sprague-Dawley) model of bone defect. Results: The developed GelMA-CM composite demonstrated improved mechanical properties, biocompatibility, and osteogenic potential compared to individual GelMA or CMCS hydrogels. Incorporation of MPC facilitated the sustained release of ions which promoted osteogenic differentiation of pre-osteoblasts. In vivo results indicated accelerated bone healing in the rat bone defect model. Conclusions: The functionalized GelMA-CM composite could be a viable candidate for clinical applications in bone regeneration therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (X.W.); (X.Z.); (C.G.); (J.Y.); (J.C.)
| |
Collapse
|
30
|
Zheng Y, Cong L, Zhao L, Wang P, Xing L, Liu J, Xu H, Li N, Zhao Y, Yuan L, Shi Q, Sun X, Liang Q, Wang Y. Lymphatic platelet thrombosis limits bone repair by precluding lymphatic transporting DAMPs. Nat Commun 2025; 16:829. [PMID: 39827193 PMCID: PMC11742876 DOI: 10.1038/s41467-025-56147-8] [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: 10/21/2023] [Accepted: 01/05/2025] [Indexed: 01/22/2025] Open
Abstract
In the musculoskeletal system, lymphatic vessels (LVs), which are interdigitated with blood vessels, travel and form an extensive transport network. Blood vessels in bone regulate osteogenesis and hematopoiesis, however, whether LVs in bone affect fracture healing is unclear. Here, we investigate the lymphatic draining function at the tibial fracture sites using near-infrared indocyanine green lymphatic imaging (NIR-ICG) and discover that lymphatic drainage insufficiency (LDI) starts on day one and persists for up to two weeks following the fracture in male mice. Sufficient lymphatic drainage facilitates fracture healing in male mice. Furthermore, we identify that lymphatic platelet thrombosis (LPT) blocks the draining lymphoid sinus and LVs, causes LDI, and inhibits fracture healing in male mice, which can be rescued by a blood thinner. Moreover, unblocked lymphatic drainage decreases neutrophils and increases M2-type macrophages of the hematoma niche to support osteoblast (OB) survival and bone marrow-derived mesenchymal stem cell (BMSC) proliferation via transporting damage-associated molecular patterns (DAMPs) in male rats. Lymphatic platelet thrombolysis also benefits senile fracture healing in female mice. These findings demonstrate that LPT limits bone regeneration by impeding lymphatic transporting DAMPs. Together, these findings represent a way forward in the treatment of bone repair.
Collapse
Affiliation(s)
- Yangkang Zheng
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China
| | - Lin Cong
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Li Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Pengyu Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine and Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Luying Yuan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Xueqing Sun
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China.
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China.
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China.
| | - YongJun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China.
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China.
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China.
| |
Collapse
|
31
|
Ma K, Wang X, Pi Z, Zhang Y, Wang R. A comparative study of the minimally invasive lateral shoulder approach and deltopectoral space approach for the treatment of proximal humerus fractures. BMC Surg 2025; 25:13. [PMID: 39773187 PMCID: PMC11706203 DOI: 10.1186/s12893-024-02690-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE To compare the clinical efficacy of the minimally invasive lateral shoulder approach and deltopectoral space approach in the treatment of proximal humerus fractures. METHODS The clinical data of 95 patients with proximal humerus fractures admitted to the hospital from June 2018 to June 2023 were retrospectively collected. Forty-four patients were treated with a minimally invasive lateral shoulder approach (study group), and 51 patients were treated with a deltopectoral space approach (control group). The baseline data (age, sex, mechanism of injury, preoperative Neer classification, and time from injury to surgery), operation time, intraoperative blood loss, incision length, fracture healing time, and postoperative complications were compared between these two groups. The VAS score, shoulder range of motion (ROM) score, and Constant-Murley score were used to evaluate the shoulder joint function of the two groups one year after surgery. RESULTS There were no significant differences in operation time, blood loss, incision length or fracture healing time between the two groups (P > 0.05). The incidence of postoperative complications in the study group was significantly lower than that in the control group, and the difference between the groups was statistically significant (P < 0.05). There was no significant difference in shoulder joint function or VAS score between the two groups one year after surgery (P > 0.05). CONCLUSION The treatment of proximal humerus fractures via the lateral shoulder approach is minimally invasive and can reduce the occurrence of complications such as ischemic necrosis of the humerus head, relieve shoulder pain in the short term, and restore good shoulder function. Therefore, given the strict grasp of indications and familiarity with surgical operations, the minimally invasive lateral shoulder approach for the treatment of proximal humeral fractures is safe and effective and is worth promoting and applying in clinical practice.
Collapse
Affiliation(s)
- Kunpeng Ma
- First Clinical School of Medicine, Southern Medical University, Guangzhou, China
- Department of Orthopedics, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, People's Republic of China
| | - Xinyu Wang
- Department of Orthopedics, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, People's Republic of China
| | - Zhilong Pi
- Department of Orthopedics, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, People's Republic of China
| | - Ying Zhang
- Department of Orthopedics, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, People's Republic of China.
| | - Renkai Wang
- Department of Orthopedics, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, People's Republic of China.
| |
Collapse
|
32
|
Chen Q, Su Y, Yang Z, Lin Q, Ke Y, Xing D, Li H. Bibliometric mapping of mesenchymal stem cell therapy for bone regeneration from 2013 to 2023. Front Med (Lausanne) 2025; 11:1484097. [PMID: 39835103 PMCID: PMC11743382 DOI: 10.3389/fmed.2024.1484097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have shown significant potential in bone regeneration and regenerative medicine in recent years. With the advancement of tissue engineering, MSCs have been increasingly applied in bone repair and regeneration, and their clinical application potential has grown through interdisciplinary approaches involving biomaterials and genetic engineering. However, there is a lack of systematic reviews summarizing their applications in bone regeneration. To address this gap, we analyzed the latest research on MSCs for bone regeneration published from 2013 to 2023. Using the Web of Science Core Collection, we conducted a literature search in December 2024 and employed bibliometric tools like CiteSpace and VOSviewer for a comprehensive analysis of the key research trends. Our findings focus on the development of cell engineering, highlighting the advantages, limitations, and future prospects of MSC applications in bone regeneration. These insights aim to enhance understanding of MSC-based bone regeneration, inspire new research directions, and facilitate the clinical translation of MSC research.
Collapse
Affiliation(s)
- Qianqian Chen
- Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Arthritis Clinic & Research Center, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiqi Su
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
| | - Zhen Yang
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
| | - Qiyuan Lin
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
| | - Yan Ke
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| | - Dan Xing
- Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Arthritis Clinic & Research Center, Zhejiang Chinese Medical University, Hangzhou, China
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| | - Hui Li
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| |
Collapse
|
33
|
Açan AE, Aydın ME, Bulmuş Ö, Özcan E, Karakılıç A, Turan G, Reşit Buğra Hüsemoğlu RBHRBH. The effects of gabapentin and pregabalin on fracture healing: A histological, radiological, and biomechanical analysis. Jt Dis Relat Surg 2025; 36:200-209. [PMID: 39719918 PMCID: PMC11734844 DOI: 10.52312/jdrs.2025.2042] [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: 10/29/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
OBJECTIVES This study evaluated the impact of different doses of gabapentin and pregabalin on fracture healing in a rat femoral shaft model, with histological, radiological, and biomechanical assessments. MATERIALS AND METHODS Seventy male Wistar albino rats were divided into five groups: control, low-dose gabapentin (GBP-L, 300 mg/day), high-dose gabapentin (GBP-H, 3600 mg/day), low-dose pregabalin (PRG-L, 150 mg/day), and high-dose pregabalin (PRG-H, 600 mg/day), based on human equivalent doses. Bilateral femoral fractures were induced; the right femurs were prepared for radiological examination using microtomography, followed by histological analysis, whereas the left femurs were allocated for biomechanical testing. Drug administration began three weeks preoperatively and continued until sacrifice at either two or four weeks. Histological assessments included inflammation and transformation scoring and microtomography-measured callus volume. Biomechanical testing assessed maximum force and stiffness. RESULTS At the fourth week, inflammation levels were significantly higher in the GBP-H, PRG-L, and PRG-H groups compared to control (p<0.01, p<0.05, and p<0.01), while transformation scores were significantly lower in these groups (p<0.01, p<0.05, and p<0.001). Low-dose pregabalin showed a borderline transformation difference (p=0.051). Microtomography analysis showed that the GBP-H group had significantly reduced callus volume versus control by the second week (p<0.01), persisting at a lower significance by week four (p<0.05). By the fourth week, PRG-H also had reduced callus volume (p<0.05). Maximum force values by the fourth week were significantly lower in the GBP-L, GBP-H, and PRG-H groups compared to control (p<0.05 for GBP-L; p<0.01 for GBP-H and PRG-H). CONCLUSION These findings suggest that these drugs, particularly with their high-dose applications, may lead to prolonged inflammation and hinder fracture healing by reducing callus volume and biomechanical integrity, potentially disrupting the transition from the inflammatory to reparative phases of healing.
Collapse
Affiliation(s)
- Ahmet Emrah Açan
- Balıkesir Üniversitesi Tıp Fakültesi Ortopedi ve Travmatoloji Anabilim Dalı, 10185 Altıeylül, Balıkesir, Türkiye.
| | | | | | | | | | | | | |
Collapse
|
34
|
Sarkar A, Gallo MC, Bell JA, Mayfield CK, Ball JR, Ayad M, Lechtholz-Zey E, Chang SW, Sugiyama O, Evseenko D, Lieberman JR. Ex Vivo Regional Gene Therapy Compared to Recombinant BMP-2 for the Treatment of Critical-Size Bone Defects: An In Vivo Single-Cell RNA-Sequencing Study. Bioengineering (Basel) 2025; 12:29. [PMID: 39851303 PMCID: PMC11762083 DOI: 10.3390/bioengineering12010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025] Open
Abstract
Ex vivo regional gene therapy is a promising tissue-engineering strategy for bone regeneration: osteogenic mesenchymal stem cells (MSCs) can be genetically modified to express an osteoinductive stimulus (e.g., bone morphogenetic protein-2), seeded onto an osteoconductive scaffold, and then implanted into a bone defect to exert a therapeutic effect. Compared to recombinant human BMP-2 (rhBMP-2), which is approved for clinical use, regional gene therapy may have unique benefits related to the addition of MSCs and the sustained release of BMP-2. However, the cellular and transcriptional mechanisms regulating the response to these two strategies for BMP-2 mediated bone regeneration are largely unknown. Here, for the first time, we performed single-cell RNA sequencing (10x Genomics) of hematoma tissue in six rats with critical-sized femoral defects that were treated with either regional gene therapy or rhBMP-2. Our unbiased bioinformatic analysis of 2393 filtered cells in each group revealed treatment-specific differences in their cellular composition, transcriptional profiles, and cellular communication patterns. Gene therapy treatment induced a more robust chondrogenic response, as well as a decrease in the proportion of fibroblasts and the expression of profibrotic pathways. Additionally, gene therapy was associated with an anti-inflammatory microenvironment; macrophages expressing canonical anti-inflammatory markers were more common in the gene therapy group. In contrast, pro-inflammatory markers were more highly expressed in the rhBMP-2 group. Collectively, the results of our study may offer insights into the unique pathways through which ex vivo regional gene therapy can augment bone regeneration compared to rhBMP-2. Furthermore, an improved understanding of the cellular pathways involved in segmental bone defect healing may allow for the further optimization of regional gene therapy or other bone repair strategies.
Collapse
Affiliation(s)
- Arijita Sarkar
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Matthew C. Gallo
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Jennifer A. Bell
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Cory K. Mayfield
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Jacob R. Ball
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Mina Ayad
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Elizabeth Lechtholz-Zey
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Stephanie W. Chang
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Jay R. Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
35
|
Burgan J, Rahmati M, Lee M, Saiz AM. Innate immune response to bone fracture healing. Bone 2025; 190:117327. [PMID: 39522707 DOI: 10.1016/j.bone.2024.117327] [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: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The field of osteoimmunology has primarily focused on fracture healing in isolated musculoskeletal injuries. The innate immune system is the initial response to fracture, with inflammatory macrophages, cytokines, and neutrophils arriving first at the fracture hematoma, followed by an anti-inflammatory phase to begin the process of new bone formation. This review aims to first discuss the current literature and knowledge gaps on the immune responses governing single fracture healing by encompassing the individual role of macrophages, neutrophils, cytokines, mesenchymal stem cells, bone cells, and other immune cells. This paper discusses the interactive effects of these cellular responses underscoring the field of osteoimmunology. The critical role of the metabolic environment in guiding the immune system properties will be highlighted along with some effective therapeutics for fracture healing in the context of osteoimmunology. However, compared to isolated fractures, which frequently heal well, long bone fractures in over 30 % of polytrauma patients exhibit impaired healing. Clinical evidence suggests there may be distinct physiologic and inflammatory pathways altered in polytrauma resulting in nonunion. Nonunion is associated with worse patient outcomes and increased societal healthcare costs. The dysregulated immunomodulatory/inflammatory response seen in polytrauma may lead to this increased nonunion rate. This paper will investigate the differences in immune response between isolated and polytrauma fractures. Finally, future directions for fracture studies are explored with consideration of the emerging roles of newly discovered immune cell functions in fracture healing, the existing challenges and conflicting results in the field, the translational potential of these studies in clinic, and the more complex nature of polytrauma fractures that can alter cell functions in different tissues.
Collapse
Affiliation(s)
- Jane Burgan
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maryam Rahmati
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109, Blindern, NO-0317 Oslo, Norway
| | - Mark Lee
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA
| | - Augustine Mark Saiz
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA.
| |
Collapse
|
36
|
Banazadeh M, Ilaghi M, Abadi B, Joushi S, Pishbin E, Dabiri S, Ramezani Farani M, Rahi A, Mostafavi E, Zare I. Chitosan nanoparticles-hydrogel composites for biomedical applications. FUNDAMENTALS AND BIOMEDICAL APPLICATIONS OF CHITOSAN NANOPARTICLES 2025:633-678. [DOI: 10.1016/b978-0-443-14088-4.00010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
37
|
Tong X, Shen X, Lin Z, Zhou R, Han Y, Zhu L, Huang S, Ma J, Li Y, Wen C, Lin J. In situ phosphorus-modified Mg 2Ge/Zn-Cu composite with improved mechanical, degradation, biotribological properties, and in vitro and in vivo osteogenesis and osteointegration performance for biodegradable bone-implant applications. Bioact Mater 2025; 43:491-509. [PMID: 40115880 PMCID: PMC11923437 DOI: 10.1016/j.bioactmat.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 08/13/2024] [Accepted: 09/18/2024] [Indexed: 03/23/2025] Open
Abstract
Zinc (Zn)-based composites are promising biodegradable bone-implant materials because of their good biocompatibility, processability, and biodegradability. Nevertheless, the low interfacial bonding strength, coordinated deformation capacity, and mechanical strength of current Zn-based composites hinder their clinical application. In this study, we developed a biodegradable in situ 4Mg2Ge/Zn-0.3Cu-0.05P composite (denoted ZMGCP) via phosphorus (P) modification and hot-rolling for bone-implant applications. The mechanical properties, corrosion behavior, biotribological performance, in vitro cytocompatibility and osteogenic differentiation, and in vivo osteogenesis and osteointegration of the as-cast (AC) and hot-rolled (HR) ZMGCP samples were systematically evaluated and compared to those of 4Mg2Ge/Zn-0.3Cu (denoted ZMGC). The primary and eutectic reinforcement Mg2Ge phases formed during solidification were refined after P modification and hot-rolling. The HR ZMGCP exhibited the best tensile properties among all the samples with an ultimate tensile strength of 288.9 MPa, a yield strength of 194.5 MPa, and an elongation of 17.7 %. The HR ZMGCP showed the lowest corrosion rate of 336 μm/a, 186 μm/a, and 61.7 μm/a as measured by potentiodynamic polarization, electrochemical impedance spectroscopy, and immersion testing, respectively, among all the samples in Hanks' solution. The HR ZMGCP also showed higher biotribological resistance than its ZMGC counterpart. The HR ZMGCP exhibited the highest in vitro cytocompatibility, the best osteogenesis capability and angiogenesis property among the HR samples of pure Zn, ZMGC, and ZMGCP. Furthermore, the HR ZMGCP displayed complete in vivo biocompatibility, osteogenesis, osteointegration capability, and an appropriate degradation rate, showing significant potential for a biodegradable bone-implant material.
Collapse
Affiliation(s)
- Xian Tong
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
- School of Materials Science and Engineering, Xiangtan University, Xiangtan, 411105, China
| | - Xinkun Shen
- Science and Education Division, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital), Wenzhou, 325016, China
| | - Zhiqiang Lin
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Runqi Zhou
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering, Higher Education and Stomatological Hospital, Chongqing Medical University, Chongqing, 401174, China
| | - Yue Han
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Li Zhu
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jianfeng Ma
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yuncang Li
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Cuie Wen
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Jixing Lin
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| |
Collapse
|
38
|
Liang W, Zhou C, Liu X, Xie Q, Xia L, Liu L, Bao W, Lin H, Xiong X, Zhang H, Zheng Z, Zhao J. Current status of nano-embedded growth factors and stem cells delivery to bone for targeted repair and regeneration. J Orthop Translat 2025; 50:257-273. [PMID: 39902262 PMCID: PMC11788687 DOI: 10.1016/j.jot.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Bone-related diseases like osteoarthritis and osteoporosis impact millions globally, affecting quality of life. Osteoporosis considerably enhances the probability of bone fractures of the wrist, hip, and spine. Enhancement and acceleration of functional bone development can be achieved through the sustained delivery of growth factors (GFs) and cells in biomaterial carriers. The delivery of bioactive compounds in a targeted, spatiotemporal way that most closely resembles the natural defect repair process can be achieved by designing the carrier system with established release kinetics. Furthermore, the carrier can serve as a substrate that mimics the extracellular matrix, facilitating osteoprogenitor cell infiltration and growth for integrative tissue healing. In this report, we explore the significance of GFs within the realm of bone and cartilage tissue engineering, encompassing their encapsulation and delivery methodologies, the kinetics of release, and their amalgamation with biomaterials and stem cells (SCs) to facilitate the mending of bone fractures. Moreover, the significance of GFs in evaluating the microenvironment of bone tissue through reciprocal signaling with cells and biomaterial scaffolds is emphasized which will serve as the foundation for prospective advances in bone and cartilage tissue engineering as well as therapeutic equipment. Nanoparticles are being used in regenerative medicine to promote bone regeneration and repair by delivering osteoinductive growth factors like BMP-2, VEGF, TGF-β. These nanocarriers allow controlled release, minimizing adverse effects and ensuring growth factors are concentrated at the injury site. They are also mixed with mesenchymal stem cells (MSCs) to improve their engraftment, differentiation, and survival. This approach is a key step in developing multi-model systems that more efficiently facilitate bone regeneration. Researchers are exploring smart nanoparticles with immunomodulatory qualities to improve bonre regeneration and reduce inflammation in injury site. Despite promising preclinical results, challenges include cost management, regulatory approval, and long term safety. However, incorporating stem cell transport and growth factors in nanoparticles could revolutionize bone regeneration and offer more personalized therapies for complex bone disorders and accidents. The translational potential of this article Stem cell transport and growth factors encapsulated in nanoparticles are becoming revolutionary methods for bone regeneration and repair. By encouraging stem cells to develop into osteoblasts, osteoinductive GFs like BMP-2, VEGF, and TGF-β can be delivered under control due to nanomaterials like nanoparticles, nanofibers, and nanotubes. By ensuring sustained release, these nanocarriers lessen adverse effects and enhance therapeutic results. In order to prove their survival and development, MCSs, which are essential for bone regeneration, are mixed with nanoparticles, frequently using scaffolds that resemble the ECM of bone. Furthermore, by adjusting to the injured environment and lowering inflammation, immunomodulatory nanostructures and stimuli-responsive nanomaterials can further maximize. While there are still shotcomings to overcome, including managing expenses, negotiating regulatory processes, and guaranteeing long-term safety, this method promises to outperform traditional bone grafting by providing quicker, more individualized, and more efficient treatments. Nano-embedded growth factors and stem cell technologies have the potential to revolutionize orthopedic therapy and significantly enhance patient outcomes with further research.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, China
| | - Xiankun Liu
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| |
Collapse
|
39
|
Pazhamannil RV, Alkhedher M. Advances in additive manufacturing for bone tissue engineering: materials, design strategies, and applications. Biomed Mater 2024; 20:012002. [PMID: 39662052 DOI: 10.1088/1748-605x/ad9dce] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/11/2024] [Indexed: 12/13/2024]
Abstract
The growing annual demand for bone grafts and artificial implants emphasizes the need for effective solutions to repair or replace injured bones. Additive manufacturing technology offers unique merits for advancing bone tissue engineering (BTE), enabling the creation of scaffolds and implants with customized shapes and designs, interconnected architecture, controlled mechanical properties and compositions, and broadening its range of applications. It overcomes the limitations of traditional manufacturing methods such as electrospinning, salt leaching, freeze drying, solvent casting etc. This review highlights additive manufacturing technologies and their applications in BTE, as well as materials and scaffold architectures to widen the potential of the biomedical sector. The selection of optimal printing methods for BTE requires careful consideration of the advantages and disadvantages against the needs for degradation, strength, and biocompatibility. Material extrusion and powder bed fusion techniques are the most widely used additive manufacturing processes in BTE. The comprehensive review also revealed that parametric designs such as triply periodic minimal surface (TPMS) and Voronoi hold better characteristics for their application in BTE. Voronoi designs exhibit exceptional randomness whereas TPMS structures feature high permeability with continuous surfaces. Topology optimized and gradient models exhibited superior physical and mechanical properties compared to uniform lattices. Future research should focus on the development of novel biomaterials, multi-material printing, assessing long-term impacts, and enhancing 3D printing technologies.
Collapse
Affiliation(s)
- Ribin Varghese Pazhamannil
- Mechanical and Industrial Engineering Department, Abu Dhabi University, PO 59911 Abu Dhabi, United Arab Emirates
| | - Mohammad Alkhedher
- Mechanical and Industrial Engineering Department, Abu Dhabi University, PO 59911 Abu Dhabi, United Arab Emirates
| |
Collapse
|
40
|
Reeves J, Tournier P, Becquart P, Carton R, Tang Y, Vigilante A, Fang D, Habib SJ. Rejuvenating aged osteoprogenitors for bone repair. eLife 2024; 13:RP104068. [PMID: 39692737 DOI: 10.7554/elife.104068] [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: 12/19/2024] Open
Abstract
Aging is marked by a decline in tissue regeneration, posing significant challenges to an increasingly older population. Here, we investigate age-related impairments in calvarial bone healing and introduce a novel two-part rejuvenation strategy to restore youthful repair. We demonstrate that aging negatively impacts the calvarial bone structure and its osteogenic tissues, diminishing osteoprogenitor number and function and severely impairing bone formation. Notably, increasing osteogenic cell numbers locally fails to rescue repair in aged mice, identifying the presence of intrinsic cellular deficits. Our strategy combines Wnt-mediated osteoprogenitor expansion with intermittent fasting, which leads to a striking restoration of youthful levels of bone healing. We find that intermittent fasting improves osteoprogenitor function, benefits that can be recapitulated by modulating NAD+-dependent pathways or the gut microbiota, underscoring the multifaceted nature of this intervention. Mechanistically, we identify mitochondrial dysfunction as a key component in age-related decline in osteoprogenitor function and show that both cyclical nutrient deprivation and Nicotinamide mononucleotide rejuvenate mitochondrial health, enhancing osteogenesis. These findings offer a promising therapeutic avenue for restoring youthful bone repair in aged individuals, with potential implications for rejuvenating other tissues.
Collapse
Affiliation(s)
- Joshua Reeves
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Pierre Tournier
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Pierre Becquart
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Robert Carton
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Yin Tang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Alessandra Vigilante
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Dong Fang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Shukry J Habib
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
41
|
Khodaei A, Nawaz Q, Zhu Z, Amin Yavari S, Weinans H, Boccaccini AR. Biomolecule and Ion Releasing Mesoporous Nanoparticles: Nonconvergent Osteogenic and Osteo-immunogenic Performance. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67491-67503. [PMID: 39576881 DOI: 10.1021/acsami.4c17540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
Immune-involved cell communications have recently been introduced as key role players in the fate of mesenchymal stem cells in making bone tissue. In this study, a drug delivery system for bone (re)generation based on copper-doped mesoporous bioactive glass nanoparticles (BGNPs) was developed to codeliver copper as a biologically active ion and icariin as an anti-inflammatory agent. This design was based on temporal inflammation fluctuations from proinflammatory to anti-inflammatory during bone generation. Three in vitro models were performed with human mesenchymal stem cells (hMSCs) to verify the osteo-immunomodulatory effects of released copper ions and icariin: nonstimulated, co-conditioned with macrophage medium and co-cultured with macrophages. Both icariin and copper showed increased levels of alkaline phosphatase activation, indicating a direct osteogenic effect. Copper-doped BGNPs showed the highest increase of osteo-immunogenic properties in a mineralization assay and also induced short-term inflammation. However, the mineralization dropped in copper doped BGNPs after loading with icariin due to copper-icariin chelate formation and inhibition of the early inflammatory phase in the immune-stimulated in vitro models. In the absence of copper, the direct osteogenic properties of icariin overtook its osteo-immunogenic inhibition and increased calcification. Overall, BGNPs doped with 5 mol % copper and no icariin showed the highest bone-forming capacity.
Collapse
Affiliation(s)
- Azin Khodaei
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
- Department of Orthopedics, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands
| | - Qaisar Nawaz
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Zhengqing Zhu
- Department of Orthopedics, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopedics, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands
| | - Aldo R Boccaccini
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| |
Collapse
|
42
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
43
|
Chen G, Qu B, Liu P, Zhang Z. Association between modified dietary inflammation index score and lumbar vertebrae bone mineral density in patients with hypertension: data from NHANES-a population-based study. Nutr Metab (Lond) 2024; 21:102. [PMID: 39633453 PMCID: PMC11616204 DOI: 10.1186/s12986-024-00877-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The modified Dietary Inflammation Index Score (M-DIS) is a scoring system used to quantify the inflammatory effects of nutrients and foods. Inflammation may affect Bone Mineral Density (BMD) and increase the risk of osteoporosis and fractures. The purpose of this study was to utilize data from the National Health and Nutrition Examination Survey (NHANES) to evaluate the relationship between M-DIS and lumbar vertebrae BMD in patients with hypertension. METHODS Data from 2007 to 2008, 2009-2010, 2013-2014 and 2017-2018 NHANES cycles were collected for secondary analysis. Information provided by NHANES participants included complete dietary intake interviews and BMD measurements. M-DIS was calculated based on dietary intake interviews. Dual energy X-ray absorptiometry (DXA) was used to evaluate the average BMD of lumbar vertebrae (L1-L4). As an indicator of bone health, weighted multiple logistic regression and restricted spline analysis were utilized to study the relationship between M-DIS and lumbar vertebrae BMD in American patients with hypertension. RESULTS A total of 3864 participants aged ≥ 20 years with complete data were included in this study. The proportion of osteopenia in the lumbar spine was 7.2%. After adjusting for confounding factors, negative correlations were observed between the BMD of each vertebral and its average BMD with M-DIS. In Model 3, the relationship between mean lumbar BMD and M-DIS was β = - 0.0103 (95% CI - 0.0160 to - 0.0046, P < 0.001). Notably, L1 showed a particularly significant negative correlation with β = - 0.0120 (95% CI - 0.0172 to - 0.0067, P < 0.001), while the proportion of osteopenia was highest in the L3 vertebra, accounting for 8.3%. Higher M-DIS was positively correlated with the incidence of osteopenia (OR 0.595, 95% CI 0.371-0.965, P = 0.041). Further analyses revealed that in hypertensive patients, elevated M-DIS in women was associated with lower lumbar BMD (P for nonlinearity = 0.093), while this trend was not significant in hypertensive men. CONCLUSIONS The results of this study suggest that a higher M-DIS (pro-inflammatory diet) is significantly associated with BMD in females with hypertension. These results indicate that female with hypertension who prefer a pro-inflammatory diet may be at an increased risk of osteopenia, highlighting the necessity for tailored dietary recommendations.
Collapse
Affiliation(s)
- Guangbin Chen
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
| | - Bo Qu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China
| | - Pan Liu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, People's Republic of China.
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China.
- Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China.
| |
Collapse
|
44
|
Huang Z, Iqbal Z, Zhao Z, Liu J, Alabsi AM, Shabbir M, Mahmood A, Liang Y, Li W, Deng Z. Cellular crosstalk in the bone marrow niche. J Transl Med 2024; 22:1096. [PMID: 39627858 PMCID: PMC11613879 DOI: 10.1186/s12967-024-05900-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 11/19/2024] [Indexed: 12/08/2024] Open
Abstract
The bone marrow niche is a special microenvironment that comprises elements, including hematopoietic stem cells, osteoblasts, and endothelial cells, and helps maintain their characteristic functions. Here, we elaborate on the crosstalk between various cellular components, hematopoietic stem cells, and other cells in the bone marrow niche. We further explain the mechanism of preserving equilibrium in the bone marrow niche, which is crucial for the directional regulation of bone reconstruction and repair. Additionally, we elucidate the intercommunication among osteocytes, the regulation of osteoblast maturation and activation by lymphocytes, the deficiency of megakaryocytes that can markedly impair osteoblast formation, and the mechanism of interaction between macrophages and mesenchymal stem cells in the bone marrow niche. Finally, we discussed the new immunotherapies for bone tumors in the BM niche. In this review, we aimed to provide a candid overview of the crosstalk among bone marrow niche cells and to highlight new concepts underlying the unknown mechanisms of hematopoiesis and bone reconstruction. Thus, this review may provide a more comprehensive understanding of the role of these niche cells in improving hematopoietic function and help identify their therapeutic potential for different diseases in the future.
Collapse
Affiliation(s)
- Zeqi Huang
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), 3002 Sungang West Road, Shenzhen, 518000, China
| | - Zoya Iqbal
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), 3002 Sungang West Road, Shenzhen, 518000, China
| | - Zhe Zhao
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), 3002 Sungang West Road, Shenzhen, 518000, China
| | - Jianquan Liu
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), 3002 Sungang West Road, Shenzhen, 518000, China
| | - A M Alabsi
- Faculty of Dentistry, MAHSA University, Selangor, Malaysia
- School of Dentistry, Management and Science University, University Drive, Off Persiaran Olahraga, 40100 ShahAlam, Selangor, Malaysia
| | - Maryam Shabbir
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Ayesha Mahmood
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Yujie Liang
- Faculty of Dentistry, MAHSA University, Selangor, Malaysia.
- Department of Child and Adolescent Psychiatry, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, 518020, China.
| | - Wencui Li
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), 3002 Sungang West Road, Shenzhen, 518000, China.
| | - Zhiqin Deng
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), 3002 Sungang West Road, Shenzhen, 518000, China.
| |
Collapse
|
45
|
Werner NL, Van Wijck SFM. Rib Fracture Nonunion. Curr Probl Surg 2024; 61:101648. [PMID: 39647975 DOI: 10.1016/j.cpsurg.2024.101648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 12/10/2024]
Affiliation(s)
- Nicole L Werner
- University of Wisconsin School of Medicine and Public Health, Department of Surgery, Division of Acute Care Regional General Surgery, Madison, WI.
| | - Suzanne F M Van Wijck
- Trauma Research Unit Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, CA, Netherlands
| |
Collapse
|
46
|
Yang J, Xiao L, Zhang L, Luo G, Ma Y, Wang X, Zhang Y. Platelets: A Potential Factor that Offers Strategies for Promoting Bone Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:631-643. [PMID: 38482796 DOI: 10.1089/ten.teb.2024.0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Bone defects represent a prevalent category of clinical injuries, causing significant pain and escalating health care burdens. Effectively addressing bone defects is thus of paramount importance. Platelets, formed from megakaryocyte lysis, have emerged as pivotal players in bone tissue repair, inflammatory responses, and angiogenesis. Their intracellular storage of various growth factors, cytokines, and membrane protein receptors contributes to these crucial functions. This article provides a comprehensive overview of platelets' roles in hematoma structure, inflammatory responses, and angiogenesis throughout the process of fracture healing. Beyond their application in conjunction with artificial bone substitute materials for treating bone defects, we propose the potential future use of anticoagulants such as heparin in combination with these materials to regulate platelet number and function, thereby promoting bone healing. Ultimately, we contemplate whether manipulating platelet function to modulate bone healing could offer innovative ideas and directions for the clinical treatment of bone defects. Impact statement Given that 5-10% of fracture patients with delayed bone healing or even bone nonunion, this review explores the potential role of platelets in bone healing (directly/indirectly) and proposes ideas and directions for the future as to whether it is possible to promote bone healing and improve fracture healing rates by modulating platelets.
Collapse
Affiliation(s)
- Jingjing Yang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, China
| | - Lan Xiao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- School of Medicine and Dentistry, Griffith University, Queensland, Australia
| | - Lijia Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Guochen Luo
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| |
Collapse
|
47
|
Bellavia D, Costa V, De Luca A, Maglio M, Pagani S, Fini M, Giavaresi G. Vitamin D Level Between Calcium-Phosphorus Homeostasis and Immune System: New Perspective in Osteoporosis. Curr Osteoporos Rep 2024; 22:599-610. [PMID: 27734322 DOI: 10.1007/s11914-016-0331-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vitamin D is a key molecule in calcium and phosphate homeostasis; however, increasing evidence has recently shown that it also plays a crucial role in the immune system, both innate and adaptive. A deregulation of vitamin D levels, due also to mutations and polymorphisms in the genes of the vitamin D pathway, determines severe alterations in the homeostasis of the organism, resulting in a higher risk of onset of some diseases, including osteoporosis. This review gives an overview of the influence of vitamin D levels on the pathogenesis of osteoporosis, between bone homeostasis and immune system.
Collapse
Affiliation(s)
- Daniele Bellavia
- Innovative Technology Platforms for Tissue Engineering, Theranostics and Oncology, Rizzoli Orthopaedic Institute, Via Divisi, 83, 90100, Palermo, Italy
| | - Viviana Costa
- Innovative Technology Platforms for Tissue Engineering, Theranostics and Oncology, Rizzoli Orthopaedic Institute, Via Divisi, 83, 90100, Palermo, Italy
| | - Angela De Luca
- Innovative Technology Platforms for Tissue Engineering, Theranostics and Oncology, Rizzoli Orthopaedic Institute, Via Divisi, 83, 90100, Palermo, Italy
| | - Melania Maglio
- Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Stefania Pagani
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Milena Fini
- Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Gianluca Giavaresi
- Innovative Technology Platforms for Tissue Engineering, Theranostics and Oncology, Rizzoli Orthopaedic Institute, Via Divisi, 83, 90100, Palermo, Italy.
| |
Collapse
|
48
|
Zeng K, Lin Y, Liu S, Wang Z, Guo L. Applications of piezoelectric biomaterials in dental treatments: A review of recent advancements and future prospects. Mater Today Bio 2024; 29:101288. [PMID: 40018432 PMCID: PMC11866170 DOI: 10.1016/j.mtbio.2024.101288] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 03/01/2025] Open
Abstract
Piezoelectric biomaterials have attracted considerable attention in dental medicine due to their unique ability to convert mechanical force into electricity and catalyze reactions. These materials demonstrate biocompatibility, high bioactivity, and stability, making them suitable for applications such as tissue regeneration, caries prevention, and periodontal disease treatment. Despite their significant potential, the clinical application of these materials in treating oral diseases remains limited, facing numerous challenges in clinical translation. Therefore, further research and data are crucial to advance their application in dentistry. The review emphasizes the transformative impact of multifunctional piezoelectric biomaterials on enhancing dental therapies and outlines future directions for their integration into oral healthcare practices.
Collapse
Affiliation(s)
- Kaichen Zeng
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yifan Lin
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shirong Liu
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziyan Wang
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lvhua Guo
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
49
|
Murayama M, Shinohara I, Toya M, Susuki Y, Lee ML, Young B, Gao Q, Chow SKH, Goodman SB. T cells and macrophages jointly modulate osteogenesis of mesenchymal stromal cells. J Biomed Mater Res A 2024; 112:2202-2209. [PMID: 38963690 DOI: 10.1002/jbm.a.37771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/12/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Approximately 5%-10% of fractures go on to delayed healing and nonunion, posing significant clinical, economic, and social challenges. Current treatment methods involving open bone harvesting and grafting are associated with considerable pain and potential morbidity at the donor site. Hence, there is growing interest in minimally invasive approaches such as bone marrow aspirate concentrate (BMAC), which contains mesenchymal stromal cells (MSCs), macrophages (Mφ), and T cells. However, the use of cultured or activated cells for treatment is not yet FDA-approved in the United States, necessitating further exploration of optimal cell types and proportions for effective bone formation. As our understanding of osteoimmunology advances, it has become apparent that factors from anti-inflammatory Mφ (M2) promote bone formation by MSCs. Additionally, M2 Mφ promote T helper 2 (Th2) cells and Treg cells, both of which enhance bone formation. In this study, we investigated the interactions among MSCs, Mφ, and T cells in bone formation and explored the potential of subsets of BMAC. Coculture experiments were conducted using primary MSCs, Mφ, and CD4+ T cells at specific ratios. Our results indicate that nonactivated T cells had no direct influence on osteogenesis by MSCs, while coculturing MSCs with Mφ and T cells at a ratio of 1:5:10 positively impacted bone formation. Furthermore, higher numbers of T cells led to increased M2 polarization and a higher proportion of Th2 cells in the early stages of coculture. These findings suggest the potential for enhancing bone formation by adjusting immune and mesenchymal cell ratios in BMAC. By understanding the interactions and effects of immune cells on bone formation, we can develop more effective strategies and protocols for treating bone defects and nonunions. Further studies are needed to investigate these interactions in vivo and explore additional factors influencing MSC-based therapies.
Collapse
Affiliation(s)
- Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yosuke Susuki
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Max L Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Bill Young
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
50
|
Qu D, Xiang J, Tian J, Zhang S, Li L, Zhou C. Enhancing bone repair efficiency through synergistic modification of recombinant human collagen onto PLLA membranes. Int J Biol Macromol 2024; 283:137631. [PMID: 39557267 DOI: 10.1016/j.ijbiomac.2024.137631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Given the exponential growth of the recombinant human collagen market, it is paramount to devise a robust and straightforward design strategy aimed at preserving the remarkable biological activity of recombinant human collagen while endowing it with tailored mechanical properties and stable morphologies. This innovative approach stands to broaden its applicability in hard tissue repair endeavors. Our study employed a synergistic approach of alkali hydrolysis and Schiff's base chemistry to graft Type I recombinant human collagen (rhCol-I) onto poly (L-lactic acid) (PLLA) membranes, yielding PLLA-rhCol composites. In vitro evaluations substantiated that this reengineered material not only retained the biological efficacy of rhCol-I but also imparted mechanical robustness and processability ideal for bone implant applications. Notably, it exhibited superior tissue engineering attributes, fostering proliferation, adhesion, osteogenic differentiation, mineralization of bone marrow mesenchymal stem cells (BMSCs), and encouraging vascularization. In a rat model of critical-sized bone defects, PLLA-rhCol exhibited markedly enhanced bone repair efficiency over conventional PLLA bone implants, achieving a bone volume fraction (BV/TV) of up to 32.57 ± 3.77 %, while promoting angiogenesis and effectively mitigating inflammatory cell infiltration. This pioneering method of modifying recombinant human collagen onto the side chains of polymeric macromolecules portends broad applicability in enhancing various biocompatible, yet mechanically robust and processable polymers, thereby expanding the horizons of recombinant human collagen utilization in tissue engineering and catering to the ever-evolving market demands.
Collapse
Affiliation(s)
- Dengjian Qu
- College of Chemistry and Materials Science, Engineering Research Center of Artificial Organs and Materials, Jinan University, Guangzhou 511486, China
| | - Junxiao Xiang
- College of Chemistry and Materials Science, Engineering Research Center of Artificial Organs and Materials, Jinan University, Guangzhou 511486, China
| | - Jinhuan Tian
- College of Chemistry and Materials Science, Engineering Research Center of Artificial Organs and Materials, Jinan University, Guangzhou 511486, China
| | - Shuyun Zhang
- Guangdong Police College, Guangzhou, Guangdong 510440, China.
| | - Lihua Li
- College of Chemistry and Materials Science, Engineering Research Center of Artificial Organs and Materials, Jinan University, Guangzhou 511486, China.
| | - Changren Zhou
- College of Chemistry and Materials Science, Engineering Research Center of Artificial Organs and Materials, Jinan University, Guangzhou 511486, China
| |
Collapse
|