1
|
Gai T, Zhang H, Hu Y, Li R, Wang J, Chen X, Wang J, Chen Z, Jing Y, Wang C, Bai L, Wang X, Su J. Sequential construction of vascularized and mineralized bone organoids using engineered ECM-DNA-CPO-based bionic matrix for efficient bone regeneration. Bioact Mater 2025; 49:362-377. [PMID: 40144795 PMCID: PMC11937690 DOI: 10.1016/j.bioactmat.2025.02.033] [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: 01/02/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Given the limitations of allogeneic and artificial bone grafts, bone organoids have attracted extensive attention for their physiological properties that closely resemble natural bone, offering great potential to bone reconstruction for critical-sized bone defects. Although early-stage bone organoids such as osteo-callus organoids and woven bone organoids have been reported, functional bone organoids with vascularization and mineralization are currently unavailable due to the lack of bone-mimicking matrix and dynamic culture systems suitable for the long-term cultivation of mature bone organoids. Herein, a novel engineered bionic matrix hydrogels with multifunctional components and double network structure are developed by incorporating calcium phosphate oligomers (CPO) into a combination of bone-derived decellularized extracellular matrix (ECM) and salmon-derived deoxyribonucleic acid (DNA) via photo-crosslinking and dynamic self-assembly strategies. This kind of bionic matrix hydrogels facilitate recruitment, proliferation, osteogenesis and angiogenesis of bone marrow mesenchymal stromal cells (BMSCs). More importantly, vascularized and mineralized bone organoids are sequentially constructed using BMSCs-loaded engineered bionic matrix hydrogels via in vitro dynamic culture and in vivo heterotopic ossification. Meanwhile, this kind of engineered bionic matrix are capable of achieving efficient bone repair for cranial defect. These findings suggest that engineered bionic matrix hydrogels combined with such dynamic culture system, providing a promising strategy for functional bone organoids construction.
Collapse
Affiliation(s)
- Tingting Gai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ruiyang Li
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jian Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jianhua Wang
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhenhua Chen
- Yantai Zhenghai Bio-tech Co., Ltd, Yantai, 264006, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Chenglong Wang
- Yantai Zhenghai Bio-tech Co., Ltd, Yantai, 264006, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiuhui Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
2
|
He X, Wang R, Liu X, Peng R, Zhou B, Wang L, Wei X, Wang S, Bai J, Feng Q, Zhou F, Liu H, Fan Y. Customizable Silk Fibroin-Based Hydrogel Fibrous Scaffold for On-Demand Multifaceted Tissue Repair. ACS NANO 2025; 19:20841-20862. [PMID: 40448661 DOI: 10.1021/acsnano.5c03283] [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: 06/02/2025]
Abstract
Hydrogel scaffolds represent an attractive tool for tissue repair. However, targeted tissue repair requires a specific shape and biological function design, and most natural-protein-based hydrogel scaffolds are predominantly confined to specific tissue repair applications. Here, we developed a versatile structural biomimetic natural protein platform through synergistic electrospinning, photopolymerization, and metal-coordination strategies. By integrating methacrylated silk fibroin (SFMA) with acrylated bisphosphonates (AcBP), we developed a dynamically functionalizable matrix that enables (1) customizable shape control via tunable electrospinning collectors and (2) on-demand biological function customization through metal-ion chelation. As a proof of concept, we demonstrate this platform's scenario-specific therapeutic efficacy: (i) Mg2+-functionalized membranes (S-LB-Mg) that orchestrate angiogenic-osteogenic coupling in critical-sized calvarial defects, (ii) Ag+-integrated dressing (S-LB-Ag) enabling bacterial eradication via a nonantibiotic mechanism and accelerating infected wound closure, and (iii) Zn2+-loaded conduits (S-LB-Zn) that drive macrophage M2 polarization to enhance peripheral nerve regeneration. This naturally derived protein-based platform overcomes the potential side effects associated with clinical bioactive factor/antibiotic composite scaffolds, offering a simple and customizable solution for the repair and regeneration of diverse tissues in a cost-effective yet highly effective manner. Overall, our strategy provides an alternative perspective for constructing protein-derived hydrogel microfibers with customizable functions and shapes for tissue repair applications.
Collapse
Affiliation(s)
- Xi He
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Ruideng Wang
- Department of Orthopaedics, Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, Beijing 100083, China
| | - Xuezhe Liu
- Key Laboratory of Biorheological Science and Technology Ministry of Education, Collage of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Renpeng Peng
- Department of Orthopedic Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430073, China
| | - Bikun Zhou
- Key Laboratory of Biorheological Science and Technology Ministry of Education, Collage of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Li Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing 210008, Jiangsu, P. R. China
| | - Xinbo Wei
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shuang Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Jinwu Bai
- Department of Orthopaedics, Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, Beijing 100083, China
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology Ministry of Education, Collage of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Fang Zhou
- Department of Orthopaedics, Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, Beijing 100083, China
| | - Haifeng Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
3
|
Su S, Bai J, Wang R, Gao S, Zhou R, Zhou F. A novel strategy for bone defect repair: Stromal cell-derived factor 1α sustained-release acellular fish scale scaffolds combined with injection of bone marrow mesenchymal stem cells promote bone regeneration. Mater Today Bio 2025; 32:101759. [PMID: 40270891 PMCID: PMC12017916 DOI: 10.1016/j.mtbio.2025.101759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Patients with bone defects often have weak cell vitality and differentiation ability of endogenous bone marrow mesenchymal stem cells (BMSCs), which makes bone regeneration face challenges. At present, the bone tissue engineering strategies are mainly to build grafts by loading cells on scaffolds in vitro. These strategies face many difficulties that limit their clinical application. To this end, we developed a new strategy for bone defect repair, namely chemotactic cell-free scaffolds combined with BMSCs injection. We first prepared a polydopamine-functionalized acellular fish scale scaffold that can continuously release stromal cell-derived factor 1α (SDF-1α) (termed as SDF-1α/PAFS) in vivo for at least 10 days. The study results showed that the scaffold not only has excellent mechanical properties and good biocompatibility but also has reactive oxygen scavenging activity, immunomodulation, angiogenesis, and osteogenesis. More importantly, SDF-1α/PAFS can recruit postoperatively injected BMSCs into bone defects for bone repair. We constructed the mouse cranial bone defect model, and in vivo experimental results confirmed that the strategy of combining SDF-1α/PAFS with BMSCs injection can effectively promote bone defect repair. Overall, this study provides a promising strategy for bone defect repair, with better clinical convenience and operability.
Collapse
Affiliation(s)
- Shilong Su
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Jinwu Bai
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Shan Gao
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Rubing Zhou
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, No.95 Yong'an Road, Xicheng, 100050, Beijing, China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| |
Collapse
|
4
|
Mujtaba AG, Altunay BB, Pinarbasi B, Topuz B, Yilgor P, Karakeçili A. Zr-based metal-organic framework nanocrystals improve the osteoinductivity and osteogenicity of alginate/methyl cellulose bioink. Int J Biol Macromol 2025; 313:144255. [PMID: 40381770 DOI: 10.1016/j.ijbiomac.2025.144255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Addressing critical-sized bone defects poses significant challenges due to the limitations of natural bone regeneration and conventional treatments like tissue transplantation. Bone tissue engineering and 3D bioprinting offer promising solutions by creating customized, biomimetic scaffolds. This study explores the innovative use of UiO-66 metal-organic framework (MOF) nanocrystals to enhance the osteoinductive and osteogenic properties of 3D bioprinted scaffolds. UiO-66 nanocrystals were synthesized and characterized, demonstrating uniform morphology and highly crystalline structure. These nanocrystals were then incorporated into alginate/methyl cellulose (AL/MC) hydrogel at various concentrations and print parameters were optimized based on physicochemical properties. AL/MC/UiO-66 optimized under specific conditions was then used as a bioink to 3D bioprint scaffolds including MC3T3-E1 preosteoblasts. Cell viability and osteogenic differentiation characteristics were assessed. The results demonstrated that AL/MC scaffolds with 2 % (w/v) UiO-66 nanocrystals significantly promoted osteogenic differentiation, as indicated by increased alkaline phosphatase (ALP) activity and upregulated expression of osteogenic markers (Runx-2, COLI, OCN). This study highlights the novel application of UiO-66 nanocrystals in 3D bioprinted scaffolds, presenting a promising strategy for enhancing bone regeneration.
Collapse
Affiliation(s)
| | - Barış Burak Altunay
- Department of Biomedical Engineering, Faculty of Engineering, Ankara University, 06135 Ankara, Turkey
| | - Begum Pinarbasi
- Department of Biomedical Engineering, Faculty of Engineering, Ankara University, 06135 Ankara, Turkey
| | - Berna Topuz
- Department of Chemical Engineering, Faculty of Engineering, Ankara University, 06100, Tandoğan, Ankara, Turkey
| | - Pinar Yilgor
- Department of Biomedical Engineering, Faculty of Engineering, Ankara University, 06135 Ankara, Turkey
| | - Ayşe Karakeçili
- Department of Chemical Engineering, Faculty of Engineering, Ankara University, 06100, Tandoğan, Ankara, Turkey.
| |
Collapse
|
5
|
Lei W, Wu Y, He P, Wu J, Chen J, Liu Y, Zhang H, de Bruijn JD, Bao C, Li Y, Ji P, Yuan H, Li M. Osteoclastogenesis-characterized osteoinductive biphasic calcium phosphate ceramic for bone regeneration in rabbit maxillary sinus lift. J Mater Chem B 2025; 13:5880-5897. [PMID: 40296547 DOI: 10.1039/d4tb02743d] [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: 04/30/2025]
Abstract
Calcium phosphate ceramics can be osteoinductive. Osteoinductive calcium phosphate ceramics are attractive bone substitutes because of their ability to induce bone formation in soft tissues far from osseous sites. Herein, we introduce an osteoinductive biphasic calcium phosphate (BCP) ceramic for maxillary sinus lift. Compared to protein-deprived bovine bone mineral (DBBM), BCP supported osteoclastogenesis in vitro and in vivo, induced bone formation following intramuscular implantation in FVB/NCrl (FVB) mice, and enhanced bone regeneration in the rabbit maxillary sinus lift. Our findings indicate that BCP is a promising bone substitute for bone regeneration in maxillary sinus lifts. Furthermore, the current information makes optimizing bone substitutes more convenient and effective. For instance, in vitro osteoclastogenesis evaluation of biomaterials can be used as the first screening, bone formation following non-osseous implantation as the follow-up and bone regeneration in pre-clinical bone defects as the final confirmation.
Collapse
Affiliation(s)
- Wei Lei
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
| | - Yan Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Ping He
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
| | - Jingqi Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
| | - Jingrong Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
| | - Yuxiao Liu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
| | - Hongmei Zhang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
| | - Joost D de Bruijn
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, MB Bilthoven 3723, The Netherlands.
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Yong Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
| | - Ping Ji
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
| | - Huipin Yuan
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, MB Bilthoven 3723, The Netherlands.
- Huipin Yuan's Lab, Sichuan, 610000, P. R. China
| | - Mingzheng Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, P. R. China.
| |
Collapse
|
6
|
Xu C, Long Y, Feng L, Liu L, Xu R, Hu W, Li H. Bionic Triboelectric Nanogenerator Activates the "mechano-electro-biochemical" Cascade Effect In Situ to Accelerate Critical-Sized Bone Defect Repair. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2504128. [PMID: 40394948 DOI: 10.1002/adma.202504128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/21/2025] [Indexed: 05/22/2025]
Abstract
The loss of the intrinsic "mechano-electro-biochemical" cascade effect in critical-sized bone defects (CSBDs) impedes the bone self-healing process. Traditional strategies cannot provide bionic electrical output, thereby failing to sufficiently activate the "mechano-electro-biochemical" cascade effect in situ and limiting the repair efficiency of CSBDs. Here, a bionic, self-adhesive, and biodegradable triboelectric nanogenerator (TENG) called PPCs-TENG using silk-fibroin-derived peptide (Cs)-grafted polydopamine-polyacrylamide (PPCs) as the electrode layer is fabricated. It provides bionic electrical stimulation (bio-ES) in response to the host's motion status. It reestablishes the resting potential during host rest and generates real-time biofeedback action potential during host movement. Further, it activates the "mechano-electro-biochemical" cascade effect for accelerating the repair of CSBDs. The bio-ES generated from PPCs-TENG enriches extracellular osteogenesis-related biochemical factors at the CSBD site. It also activates the intracellular mechanosensitive protein Piezo1, thereby promoting calcium signaling and intracellular mechano-transduction pathways. This activation enhances the proliferation and migration of bone marrow stem cells (BMSCs) and human umbilical vein endothelial cells (HUVECs), and promotes osteogenic differentiation in BMSCs and angiogenesis in HUVECs. In vivo tests demonstrate that PPCs-TENG significantly accelerates the repair of CSBDs in situ.
Collapse
Affiliation(s)
- Changzhen Xu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yong Long
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Lin Liu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Rongchen Xu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Weiguo Hu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Hongbo Li
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
7
|
Yu F, Zhao X, Jiang T, Ouyang N, Li P, Yang W, Zhao Z. Enhancing Osteogenesis through Bio-Inspired Recombinant Coral Protein Galaxin by Targeting Mitochondrial Metabolism and ATP Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412867. [PMID: 40056046 PMCID: PMC12061279 DOI: 10.1002/advs.202412867] [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] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/17/2025] [Indexed: 03/17/2025]
Abstract
Bio-inspired coral-derived scaffolds have been adopted for bone regeneration. However, these confront challenges such as the limited osteoinductive properties, environmental concerns, and ambiguous calcification mechanisms in coral proteins. In this study, the role of the recombinant coral protein galaxin in promoting osteogenesis is investigated. The observations reveal that recombinant galaxin regulates the mitochondrial metabolism in mouse pre-osteoblasts by interacting with the β subunit of ATP synthase. This ultimately promotes osteogenesis of pre-osteoblasts. Furthermore, galaxin is integrated with gelatin methacryloyl (GelMA) and assess the osteogenic potential of the resulting galaxin-GelMA composites in mouse mandibular defects. The observations emphasize the distinctive role of galaxin in regulating mitochondrial functionality and osteogenesis. Additionally, these provide prospective insights for further applications of bio-inspired recombinant coral proteins in regenerative medicine.
Collapse
Affiliation(s)
- Fei Yu
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesDepartment of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Xing Zhao
- Department of NephrologyKidney Research InstituteWest China Hospital of Sichuan UniversityChengdu610041China
- Institute of Biomedical EngineeringCollege of MedicineSouthwest Jiaotong UniversityChengdu610031China
| | - Ting Jiang
- Department of Pediatric DentistryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Ningjuan Ouyang
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Peilin Li
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesDepartment of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Wei Yang
- College of Polymer Science and EngineeringMed‐X Center for MaterialsSichuan UniversityChengdu610065China
| | - Zhihe Zhao
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesDepartment of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengdu610041China
| |
Collapse
|
8
|
Chen X, Weng Z, Zhang H, Jiao J, Liang J, Xu J, Wang D, Liu Q, Yan Q, Gu A. Nano-zinc oxide (nZnO) targets the AMPK-ULK1 pathway to promote bone regeneration. Stem Cell Res Ther 2025; 16:206. [PMID: 40275329 PMCID: PMC12023698 DOI: 10.1186/s13287-025-04322-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Nano-zinc oxide (nZnO) has attracted significant attention in bone tissue engineering due to its antibacterial properties, anti-inflammatory effects, biocompatibility, and chemical stability. Although numerous studies have demonstrated the enhancement of osteogenic differentiation by nZnO-modified tissue engineering materials, the underlying mechanisms remain poorly characterized. METHODS This study aimed to identify the molecular mechanisms how nZnO promoted osteogenic differentiation and bone regeneration using transcriptome analysis, drug intervention, and shRNA knockdown techniques, etc. First, the study evaluated the in vivo effects of gelatin methacryloyl (GelMA) containing nZnO on bone regeneration using a mouse calvarial defect model. The impact of nZnO exposure on the osteogenic differentiation of mesenchymal stem cells (MSCs) was then assessed. The combined treatment of nZnO and MSCs in GelMA for bone regeneration was assessed in the mouse calvarial defect model thereafter. RESULTS nZnO induced osteoblastic differentiation to promote bone regeneration. nZnO activated the AMP-dependent protein kinase (AMPK)-ULK1 signals to stimulate autophagosomes formation and facilitate autophagy flow, which was the essential pathway to induce osteogenic differentiation. The combined treatment of MSCs and nZnO significantly enhanced bone regeneration in calvarial defect mice. Conversely, AMPK inhibitor Compound C (C.C) reversed the effects on autophagy flow and osteogenic potentiality induced by nZnO. CONCLUSIONS These results highlight that nZnO can regulate bone regeneration by activating autophagy through the AMPK/ULK1 signaling pathway, which may provide a novel therapeutic strategy for addressing bone defects using nZnO.
Collapse
Affiliation(s)
- Xiu Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Environmental Health Risk Assessment Engineering Research Center, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhenkun Weng
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Environmental Health Risk Assessment Engineering Research Center, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
- Changzhou Second People's Hospital, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 213004, China
| | - Hongchao Zhang
- School of Medicine, Shanghai East Hospital & Institute of Gallstone Disease, Tongji University, Shanghai, Nanjing, 200120, 211166, China
| | - Jian Jiao
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Environmental Health Risk Assessment Engineering Research Center, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jingjia Liang
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Environmental Health Risk Assessment Engineering Research Center, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jin Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Environmental Health Risk Assessment Engineering Research Center, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Dongmei Wang
- Changzhou Second People's Hospital, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 213004, China
| | - Qian Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
- Jiangsu Environmental Health Risk Assessment Engineering Research Center, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Qing Yan
- Department of Neurosurgery, Children's Hospital of Nanjing Medical University, Nanjing, 211166, China.
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
- Jiangsu Environmental Health Risk Assessment Engineering Research Center, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
9
|
De Pace R, Molinari S, Mazzoni E, Perale G. Bone Regeneration: A Review of Current Treatment Strategies. J Clin Med 2025; 14:1838. [PMID: 40142646 PMCID: PMC11943102 DOI: 10.3390/jcm14061838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Bone regeneration has emerged as a critical research and clinical advancement field, fueled by the growing demand for effective treatments in orthopedics and oncology. Over the past two decades, significant progress in biomaterials and surgical techniques has led to the development of novel solutions for treating bone defects, surpassing the use of traditional autologous grafts. This review aims to assess the latest approaches in bone regeneration, including autologous, allogenic, and xenogenic grafts, naturally derived biomaterials, and innovative synthetic substitutes such as bioceramics, bioactive glasses, metals, polymers, composite materials, and other specialized applications. A comprehensive literature search was conducted on PubMed, focusing on studies published between 2019 and 2024, including meta-analyses, reviews, and systematic reviews. The review evaluated a range of bone regeneration strategies, examining the clinical outcomes, materials used, surgical techniques, and the effectiveness of various approaches in treating bone defects. The search identified numerous studies, with the inclusion criteria focused on those exploring innovative bone regeneration strategies. These studies provided valuable insights into the clinical and biological outcomes of different biomaterials and graft types. Results indicated that while advancements in synthetic and naturally derived biomaterials show promising potential, challenges remain in optimizing therapeutic strategies across diverse patient populations and clinical settings. The findings emphasize the need for an integrated approach that combines scientific research, clinical practice, and technological innovation to improve bone regeneration therapies. Further research is required to establish standardized protocols and determine the optimal application of various materials and techniques to enhance patient outcomes and the quality of care.
Collapse
Affiliation(s)
- Raffaella De Pace
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Silvia Molinari
- Industrie Biomediche Insubri SA, Via Cantonale 67, 6805 Mezzovico-Vira, Switzerland
| | - Elisa Mazzoni
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Giuseppe Perale
- Industrie Biomediche Insubri SA, Via Cantonale 67, 6805 Mezzovico-Vira, Switzerland
- Faculty of Biomedical Sciences, University of Southern Switzerland (USI), Via G. Buffi 13, 6900 Lugano, Switzerland
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| |
Collapse
|
10
|
Zhang L, Su L, Wu L, Zhou W, Xie J, Fan Y, Zhou X, Zhou C, Cui Y, Sun J. Versatile hydrogels prepared by microfluidics technology for bone tissue engineering applications. J Mater Chem B 2025; 13:2611-2639. [PMID: 39876639 DOI: 10.1039/d4tb02314e] [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: 01/30/2025]
Abstract
Bone defects are a prevalent issue resulting from various factors, such as trauma, degenerative diseases, congenital disabilities, and the surgical removal of tumors. Current methods for bone regeneration have limitations. In this context, the fusion of tissue engineering and microfluidics has emerged as a promising strategy in the field of bone regeneration. This study describes the classification of microfluidic devices based on the nature of flow and channel type, as well as the materials and techniques required. An overview of microfluidic methods used to prepare hydrogels and the advantages of using these hydrogels in bone tissue engineering (BTE) combining several basic elements of BTE to highlight its advantages is provided. Furthermore, this work emphasizes the benefits of using hydrogels prepared via microfluidics over conventional hydrogels in BTE because of their controlled release of cargo, they can be used for in situ injection, simplify the steps of single-cell encapsulation and have the advantages of high-throughput and precise preparation. Additionally, organ-on-a-chip models fabricated via microfluidics offer a platform for studying cell and tissue behaviors in an authentic and dynamic environment. Moreover, microfluidic devices can be utilized for noninvasive diagnosis and therapy. Finally, this paper summarizes the preclinical and clinical applications of hydrogels prepared via microfluidics for bone regeneration by focusing on their current developmental status, limitations associated with their application, and future challenges, which underscore their potential impacts on advancing regenerative medicine practices.
Collapse
Affiliation(s)
- Luyue Zhang
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Liqian Su
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Lina Wu
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Weikai Zhou
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jing Xie
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Yi Fan
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Changchun Zhou
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yujia Cui
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jianxun Sun
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
11
|
Wang Y, Zhou Y, Li K. The role of lncRNA in the differentiation of adipose-derived stem cells: from functions to mechanism. J Mol Med (Berl) 2025; 103:125-135. [PMID: 39708157 DOI: 10.1007/s00109-024-02507-8] [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: 07/04/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
Adipose-derived stem cells (ADSCs) have become one of the best seed cells widely studied and concerned in tissue engineering because of their rich sources and excellent multi-directional differentiation ability, which are expected to play a practical application role in tissue defect, osteoporosis, plastic surgery, and other fields. However, the differentiation direction of ADSCs is regulated by complex factors. Long non-coding RNAs (lncRNAs) are RNA molecules longer than 500 nucleotides that do not encode proteins and can act as signaling RNAs in response to intracellular and extracellular stimuli. Recently, accumulating evidence has revealed that lncRNAs could regulate the cell cycle and differentiation direction of ADSCs through various mechanisms, including histone modification, binding to RNA-binding proteins, and regulating the expression of miRNAs. Therefore, enriching and elucidating its mechanism of action as well as targeting lncRNAs to regulate ADSCs differentiation have potential prospects in tissue regeneration applications such as bone, blood vessels, and adipose. In this review, we summarize the role and mechanism of lncRNAs and its complexes in the multi-directional differentiation of ADSCs and discuss some potential approaches that can exert therapeutic effects on tissue defects by modulating the expression level of lncRNAs in ADSCs. Our work might provide some new research directions for the clinical applications of tissue engineering.
Collapse
Affiliation(s)
- Yujing Wang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, 410008, Hunan, China
| | - Yuxi Zhou
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, 410008, Hunan, China
| | - Kun Li
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Central South University, Changsha, 410000, China.
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Bian H, Song F, Wang S, Sun W, Hu B, Liang X, Yang H, Huang C. Matrix vesicle-inspired delivery system based on nanofibrous chitosan microspheres for enhanced bone regeneration. Mater Today Bio 2025; 30:101448. [PMID: 39866778 PMCID: PMC11762186 DOI: 10.1016/j.mtbio.2025.101448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/01/2025] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Inspired by the initial mineralization process with bone matrix vesicles (MVs), this study innovatively developed a delivery system to mediate mineralization during bone regeneration. The system comprises nanofibrous chitosan microspheres (NCM) and poly (allylamine hydrochloride)-stabilized amorphous calcium phosphate (PAH-ACP), which is thereafter referred to as NCMP. NCM is synthesized through the thermal induction of chitosan molecular chains, serving as the carrier, while PAH-ACP functions as the mineralization precursor. Additionally, the nanofibrous network of NCMP mimics the architecture of natural extracellular matrix (ECM), creating an optimal niche for the active adhesion of stem cells to its surface, exhibiting good biocompatibility, immunoregulation, and osteogenic performance. In vivo, NCMP effectively recruits cells and mineralizes collagen, modulates cell behavior and differentiation, and promotes in situ biomineralization in rat calvarial defects. These results underscore the dual efficacy of NCMP not only as an effective delivery system for mineralization precursors but also as ECM-mimicking bio-blocks, offering a promising avenue for enhancing the repair and regeneration of bone defects.
Collapse
Affiliation(s)
- Haolin Bian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Department of Stomatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Shilei Wang
- Key Laboratory of Resources and Compound of Traditional Chinese Medicine, Ministry of Education, Hubei University of Traditional Chinese Medicine, Wuhan, 430065, China
| | - Wei Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Bo Hu
- Research and Application of Regenerative Cellulose Fiber Key Laboratory of Sichuan Province, YiBin Grace Group Co., LTD, Yibin, 644000, China
| | - Xichao Liang
- Research and Application of Regenerative Cellulose Fiber Key Laboratory of Sichuan Province, YiBin Grace Group Co., LTD, Yibin, 644000, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
15
|
Sun Y, Liu J, Chen K, Zhong N, He C, Luan X, Zang X, Sun J, Cao N, Wang W, Ren Q. Polydopamine grafting polyether ether ketone to stabilize growth factor for efficient osteonecrosis repair. Sci Rep 2025; 15:3697. [PMID: 39880837 PMCID: PMC11779900 DOI: 10.1038/s41598-025-86965-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
This study examines the biocompatibility, osteogenic potential, and effectiveness of polyether ether ketone (PEEK) composites for treating osteonecrosis, seeking to establish a theoretical basis for clinical application. A range of PEEK composite materials, including sulfonated polyether ether ketone (SPEEK), polydopamine-sulfonated polyether ether ketone (SPEEK-PDA), bone-forming peptide-poly-dopamine-sulfonated polyether ether ketone (SPEEK-PDA-BFP), and vascular endothelial growth factor-poly-dopamine-sulfonated polyether ether ketone (SPEEK-PDA-VEGF), were constructed by concentrated sulfuric acid sulfonation, polydopamine modification and grafting of bioactive factors. The experiments involved adult male New Zealand rabbits aged 24-28 weeks and weighing 2.6-4 kg. The SPEEK-PDA-BFP possesses the smallest water contact angle, indicating the highest hydrophilicity, with its surface characterized by a rich density of clustered BFP particles. The SPEEK-PDA-BFP exhibits superior adhesion, proliferation, and differentiation capabilities, along with pronounced bacteriostatic effects, which are attributed to its dense particle clusters. The SPEEK-PDA-BFP facilitates the formation of regular and dense bone trabeculae. Comparative study on treating osteonecrosis with SPEEK-PDA-VEGF and SPEEK-PDA-BFP highlighted the superior formation of mature bone trabeculae and angiogenic protein CD31 around SPEEK-PDA-VEGF. The PEEK composite materials have good biocompatibility, osteogenic activity and bone repair activity. In particular, SPEEK-PDA-VEGF composite materials have the best effect on bone repair.
Collapse
Affiliation(s)
- Yi Sun
- Department of Bone Joint, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256600, China
| | - Jingyun Liu
- Department of Orthopedics, Liaocheng People's Hospital, No. 67, Dongchang West Road, Liaocheng, 252000, China
| | - Kaijia Chen
- Department of Bone Joint, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256600, China
| | - Nannan Zhong
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Chengpeng He
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Xinming Luan
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Xiaobei Zang
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Jianbo Sun
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Ning Cao
- School of Materials and Engineering, China University of Petroleum (East China), No.66, West Changjiang Road, Huangdao District, Qingdao, 266580, China
| | - Wenbo Wang
- Department of Orthopedics, The Second Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan, 250001, China
| | - Qiang Ren
- Department of Bone Joint, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256600, China.
| |
Collapse
|
16
|
Li F, Ye J, Liu P, Jiang J, Chen X. An Overview on Bioactive Glasses for Bone Regeneration and Repair: Preparation, Reinforcement, and Applications. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 39761075 DOI: 10.1089/ten.teb.2024.0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Synthetic bone transplantation has emerged in recent years as a highly promising strategy to address the major clinical challenge of bone tissue defects. In this field, bioactive glasses (BGs) have been widely recognized as a viable alternative to traditional bone substitutes due to their unique advantages, including favorable biocompatibility, pronounced bioactivity, excellent biodegradability, and superior osseointegration properties. This article begins with a comprehensive overview of the development and success of BGs in bone tissue engineering, and then focuses on their composite reinforcement systems with biodegradable metals, calcium-phosphorus (Ca-P)-based bioceramics, and biodegradable medical polymers, respectively. Moreover, the article outlines some frequently used manufacturing methods for three-dimensional BG-based bone bioscaffolds and highlights the remarkable achievements of these scaffolds in the field of bone defect repair in recent years. Lastly, based on the many potential challenges encountered in the preparation and application of BGs, a brief outlook on their future directions is presented. This review may help to provide new ideas for researchers to develop ideal BG-based bone substitutes for bone reconstruction and functional recovery.
Collapse
Affiliation(s)
- Fulong Li
- Materials Science and Engineering, School of Materials and Chemistry, University of Shanghai for Science & Technology, Shanghai, China
| | - Juelan Ye
- Biomedical Engineering, School of Health Science and Engineering, University of Shanghai for Science & Technology, Shanghai, China
| | - Ping Liu
- Materials Science and Engineering, School of Materials and Chemistry, University of Shanghai for Science & Technology, Shanghai, China
| | - Jiaqi Jiang
- Materials Science and Engineering, School of Materials and Chemistry, University of Shanghai for Science & Technology, Shanghai, China
| | - Xiaohong Chen
- Materials Science and Engineering, School of Materials and Chemistry, University of Shanghai for Science & Technology, Shanghai, China
| |
Collapse
|
17
|
Yang L, Li W, Ding X, Zhao Y, Qian X, Shang L. Biomimetic Mineralized Organic–Inorganic Hybrid Scaffolds From Microfluidic 3D Printing for Bone Repair. ADVANCED FUNCTIONAL MATERIALS 2025; 35. [DOI: 10.1002/adfm.202410927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Indexed: 01/12/2025]
Abstract
AbstractBone defect is a common clinical orthopedic disease. The trend in this field is to develop tissue engineering scaffolds with appropriately designed components and structures for bone repair. Herein, inspired by the organic and inorganic components of bone matrix and the natural biomineralization mechanism, a MgSiO3@Fe3O4 nanoparticle composite polycaprolactone (PCL) hybrid mineralized scaffold for bone repair is developed by microfluidic 3D printing. The incorporation of MgSiO3@Fe3O4 within the PCL scaffold can effectively improve the bioactivity. In addition, a biomimetic mineralized layer is prepared on the surface of the scaffold, which endowed it with unique microstructural characteristics, enhanced cell adhesion and osteogenic activity, and thus improved the bone repair performance. Owing to these advantages, both in vivo and in vitro experiments have demonstrated that the designed scaffold has outstanding biocompatibility and bone repair performance. These features indicate that the organic–inorganic biomineralized hybrid scaffold can be a potential bone graft substitute for clinical bone repair.
Collapse
Affiliation(s)
- Lei Yang
- Department of Otolaryngology Head and Neck Surgery Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health) Wenzhou Institute University of Chinese Academy of Sciences Wenzhou 325001 China
| | - Wenzhao Li
- Department of Otolaryngology Head and Neck Surgery Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health) Wenzhou Institute University of Chinese Academy of Sciences Wenzhou 325001 China
| | - Xiaoya Ding
- Department of Otolaryngology Head and Neck Surgery Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health) Wenzhou Institute University of Chinese Academy of Sciences Wenzhou 325001 China
| | - Yuanjin Zhao
- Department of Otolaryngology Head and Neck Surgery Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health) Wenzhou Institute University of Chinese Academy of Sciences Wenzhou 325001 China
| | - Xiaoyun Qian
- Department of Otolaryngology Head and Neck Surgery Nanjing Drum Tower Hospital Affiliated Hospital of Medical School Nanjing University Nanjing 210008 China
| | - Luoran Shang
- Department of Otolaryngology Head and Neck Surgery Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
- Shanghai Xuhui Central Hospital Zhongshan‐Xuhui Hospital and the Shanghai Key Laboratory of Medical Epigenetics International Co‐laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology) Institutes of Biomedical Sciences Fudan University Shanghai 200032 China
| |
Collapse
|
18
|
Fang L, Lin X, Xu R, Liu L, Zhang Y, Tian F, Li JJ, Xue J. Advances in the Development of Gradient Scaffolds Made of Nano-Micromaterials for Musculoskeletal Tissue Regeneration. NANO-MICRO LETTERS 2024; 17:75. [PMID: 39601962 PMCID: PMC11602939 DOI: 10.1007/s40820-024-01581-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
The intricate hierarchical structure of musculoskeletal tissues, including bone and interface tissues, necessitates the use of complex scaffold designs and material structures to serve as tissue-engineered substitutes. This has led to growing interest in the development of gradient bone scaffolds with hierarchical structures mimicking the extracellular matrix of native tissues to achieve improved therapeutic outcomes. Building on the anatomical characteristics of bone and interfacial tissues, this review provides a summary of current strategies used to design and fabricate biomimetic gradient scaffolds for repairing musculoskeletal tissues, specifically focusing on methods used to construct compositional and structural gradients within the scaffolds. The latest applications of gradient scaffolds for the regeneration of bone, osteochondral, and tendon-to-bone interfaces are presented. Furthermore, the current progress of testing gradient scaffolds in physiologically relevant animal models of skeletal repair is discussed, as well as the challenges and prospects of moving these scaffolds into clinical application for treating musculoskeletal injuries.
Collapse
Affiliation(s)
- Lei Fang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Xiaoqi Lin
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Ruian Xu
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Lu Liu
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yu Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Feng Tian
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| |
Collapse
|
19
|
Qi ML, Li M, Yuan K, Song E, Zhang H, Yao S. Fabrication and X-ray microtomography of sandwich-structured PEEK implants for skull defect repair. Sci Rep 2024; 14:28585. [PMID: 39562669 PMCID: PMC11577077 DOI: 10.1038/s41598-024-80103-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024] Open
Abstract
Bone defects pose a significant risk to human health. Medical polyetheretherketone (PEEK) is an excellent implant material for bone defect repair, but it faces the challenge of bone osteoconduction and osseointegration. Osteoconduction describes the process by which bone grows on the surface of the implant, while osseointegration is the stable anchoring of the implant achieved by direct contact between the bone and the implant. Bone defects repair depends on the implant's three-dimensional spatial structure, including pore size, porosity, and interconnections to a great extent. However, it is challenging to fabricate the porous structures to meet specific requirements and to characterize them without causing damage. In this study, we designed and fabricated sandwich-like PEEK implants mimicking the three-layer structures of the skull, whose defects imposes a significant burden on young adulthood and paediatric populations, and performed in-line phase-contrast synchrotron X-ray microtomography to non-destructively investigate the internal porous microstructures. The sandwich-like three-layer microstructure, comprising a dense layer, a loose layer and a dense layer in succession, exhibits structural similarity to that in a natural skull. This work demonstrated the fabrication of the sandwich-like PEEK implant that could potentially enhance osteoconduction and osseointegration. Furthermore, the interior structures and residual porogen sodium chloride particles were observed within the PEEK implant, which cannot be realized by other microscopic methods without destroying the sample. It highlights the advantages and potential of using synchrotron X-ray microtomography to analyze the structure of biomedical materials. This study provides theoretical guidance for the further design and fabrication of PEEK bone repair materials and will advance the clinical application of innovative bioactive bone repair materials.
Collapse
Affiliation(s)
- Mei-Li Qi
- School of Civil Engineering, Shandong Jiaotong University, Jinan, 250357, China
- National United Engineering Laboratory for Biomedical Material Modification, Dezhou, 251100, China
| | - Minghua Li
- School of Civil Engineering, Shandong Jiaotong University, Jinan, 250357, China
| | - Kunshan Yuan
- National United Engineering Laboratory for Biomedical Material Modification, Dezhou, 251100, China
| | - Enhui Song
- National United Engineering Laboratory for Biomedical Material Modification, Dezhou, 251100, China
| | - Haijun Zhang
- National United Engineering Laboratory for Biomedical Material Modification, Dezhou, 251100, China.
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Shengkun Yao
- Shandong Provincial Engineering and Technical Center of Light Manipulations & Shandong Provincial Key Laboratory of Optics and Photonic Device, School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China.
- Collaborative Innovation Center of Light Manipulation and Applications, Shandong Normal University, Jinan, 250358, China.
| |
Collapse
|
20
|
Ma X, Tian Y, Yang R, Wang H, Allahou LW, Chang J, Williams G, Knowles JC, Poma A. Nanotechnology in healthcare, and its safety and environmental risks. J Nanobiotechnology 2024; 22:715. [PMID: 39548502 PMCID: PMC11566612 DOI: 10.1186/s12951-024-02901-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: 07/22/2024] [Accepted: 10/03/2024] [Indexed: 11/18/2024] Open
Abstract
Nanotechnology holds immense promise in revolutionising healthcare, offering unprecedented opportunities in diagnostics, drug delivery, cancer therapy, and combating infectious diseases. This review explores the multifaceted landscape of nanotechnology in healthcare while addressing the critical aspects of safety and environmental risks associated with its widespread application. Beginning with an introduction to the integration of nanotechnology in healthcare, we first delved into its categorisation and various materials employed, setting the stage for a comprehensive understanding of its potential. We then proceeded to elucidate the diverse healthcare applications of nanotechnology, spanning medical diagnostics, tissue engineering, targeted drug delivery, gene delivery, cancer therapy, and the development of antimicrobial agents. The discussion extended to the current situation surrounding the clinical translation and commercialisation of these cutting-edge technologies, focusing on the nanotechnology-based healthcare products that have been approved globally to date. We also discussed the safety considerations of nanomaterials, both in terms of human health and environmental impact. We presented the in vivo health risks associated with nanomaterial exposure, in relation with transport mechanisms, oxidative stress, and physical interactions. Moreover, we highlighted the environmental risks, acknowledging the potential implications on ecosystems and biodiversity. Lastly, we strived to offer insights into the current regulatory landscape governing nanotechnology in healthcare across different regions globally. By synthesising these diverse perspectives, we underscore the imperative of balancing innovation with safety and environmental stewardship, while charting a path forward for the responsible integration of nanotechnology in healthcare.
Collapse
Affiliation(s)
- Xiaohan Ma
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK.
| | - Yaxin Tian
- United InnoMed (Shanghai) Limited, F/2, E-1, No.299, Kangwei Rd, Pudong District, Shanghai, China
| | - Ren Yang
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK
| | - Haowei Wang
- Centre for Precision Healthcare, UCL Division of Medicine, University College London, London, WC1E 6JF, UK
| | - Latifa W Allahou
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jinke Chang
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Division of Surgery & Interventional Science, University College London, London, NW3 2PF, UK
| | - Gareth Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jonathan C Knowles
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Med-Icine, Dankook University, Cheonan, 31116, South Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, South Korea
| | - Alessandro Poma
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK.
| |
Collapse
|
21
|
Zhang X, Chen Y, Zhou S, Liu Y, Zhu S, Jia X, Lu Z, Zhang Y, Zhang W, Ye Z, Cai B, Kong L, Liu F. RNA Coating Promotes Peri-Implant Osseointegration. ACS Biomater Sci Eng 2024; 10:7030-7042. [PMID: 38943625 PMCID: PMC11558559 DOI: 10.1021/acsbiomaterials.4c00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
In addition to transmitting and carrying genetic information, RNA plays an important abiotic role in the world of nanomaterials. RNA is a natural polyanionic biomacromolecule, and its ability to promote osteogenesis by binding with other inorganic materials as an osteogenic induction agent was discovered only recently. However, whether it can promote osseointegration on implants has not been reported. Here, we investigated the effect of the RNA-containing coating materials on peri-implant osseointegration. Total RNA extracted from rat muscle tissue was used as an osteogenic induction agent, and hyaluronic acid (HA) was used to maintain its negative charge. In simulated body fluids (SBF), in vitro studies demonstrated that the resulting material encouraged calcium salt deposition. Cytological experiments showed that the RNA-containing coating induced greater cell adhesion and osteogenic differentiation in comparison to the control. The results of animal experiments showed that the RNA-containing coating had osteoinductive and bone conduction activities, which are beneficial for bone formation and osseointegration. Therefore, the RNA-containing coatings are useful for the surface modification of titanium implants to promote osseointegration.
Collapse
Affiliation(s)
- Xiao Zhang
- College
of Life Sciences, Northwest University, Xi’an 710069, China
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Yicheng Chen
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Shanluo Zhou
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Ya Liu
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Simin Zhu
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Xuelian Jia
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Zihan Lu
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Yufan Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Wenhui Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Zhou Ye
- Applied
Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, S.A.R., China
| | - Bolei Cai
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Liang Kong
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Fuwei Liu
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| |
Collapse
|
22
|
Zheng W, Zhu Z, Hong J, Wang H, Cui L, Zhai Y, Li J, Wang C, Wang Z, Xu L, Hao Y, Cheng G, Ma S. Incorporation of small extracellular vesicles in PEG/HA-Bio-Oss hydrogel composite scaffold for bone regeneration. Biomed Mater 2024; 19:065014. [PMID: 39312942 DOI: 10.1088/1748-605x/ad7e6c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/23/2024] [Indexed: 09/25/2024]
Abstract
Stem cell derived small extracellular vesicles (sEVs) have emerged as promising nanomaterials for the repair of bone defects. However, low retention of sEVs affects their therapeutic effects. Clinically used natural substitute inorganic bovine bone mineral (Bio-Oss) bone powder lacks high compactibility and efficient osteo-inductivity that limit its clinical application in repairing large bone defects. In this study, a poly ethylene glycol/hyaluronic acid (PEG/HA) hydrogel was used to stabilize Bio-Oss and incorporate rat bone marrow stem cell-derived sEVs (rBMSCs-sEVs) to engineer a PEG/HA-Bio-Oss (PEG/HA-Bio) composite scaffold. Encapsulation and sustained release of sEVs in hydrogel scaffold can enhance the retention of sEVs in targeted area, achieving long-lasting repair effect. Meanwhile, synergistic administration of sEVs and Bio-Oss in cranial defect can improve therapeutic effects. The PEG/HA-Bio composite scaffold showed good mechanical properties and biocompatibility, supporting the growth of rBMSCs. Furthermore, sEVs enhancedin vitrocell proliferation and osteogenic differentiation of rBMSCs. Implantation of sEVs/PEG/HA-Bio in rat cranial defect model promotedin vivobone regeneration, suggesting the great potential of sEVs/PEG/HA-Bio composite scaffold for bone repair and regeneration. Overall, this work provides a strategy of combining hydrogel composite scaffold systems and stem cell-derived sEVs for the application of tissue engineering repair.
Collapse
Affiliation(s)
- Wenlong Zheng
- Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou 215028, People's Republic of China
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Zhanchi Zhu
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
- School of Nano-Technology and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Jing Hong
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
- School of Nano-Technology and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Hao Wang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China
| | - Leisha Cui
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
- School of Nano-Technology and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Yuanxin Zhai
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
- School of Nano-Technology and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Jiawei Li
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
- School of Nano-Technology and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Chen Wang
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Zhaojun Wang
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Lunshan Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China
| | - Ying Hao
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
- School of Nano-Technology and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Guangdong Institute of Semiconductor Micro-Nano Manufacturing Technology, Guangdong 528200, People's Republic of China
| | - Guosheng Cheng
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
- School of Nano-Technology and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Guangdong Institute of Semiconductor Micro-Nano Manufacturing Technology, Guangdong 528200, People's Republic of China
| | - Sancheng Ma
- Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou 215028, People's Republic of China
| |
Collapse
|
23
|
Hu X, Yang S, Zhao W, Zhang Z, Qiao L, Wu H, Su Q, Che L, Zhou K, Li K, He J. Novel multi-functional microsphere scaffold with shape memory function for bone regeneration. BIOMATERIALS ADVANCES 2024; 163:213958. [PMID: 39053385 DOI: 10.1016/j.bioadv.2024.213958] [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: 05/07/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
Irregular bone defects caused by trauma and bone diseases provide a complex implant environment for surgery. Traditional implants often fail to integrate well with the surrounding bone defect interface, therefore, developing an artificial bone scaffold that can adapt to irregular bone defect boundaries is of significant importance for bone defect repair. This study successfully utilized a shape memory ternary copolymer polylactic acid-trimethylene carbonate-hydroxyacetic acid (PLLA-TMC-GA) and dopamine-modified nano-hydroxyapatite (PHA) composite to construct a temperature-responsive bone repair scaffold (PTG/PHA), thereby enhancing the interface compatibility between the implant and the surrounding environment. The addition of PHA has effectively improved the hydrophilicity of the stent and significantly increased its mechanical strength. Furthermore, the Sodium alginate (SA) hydrogel loaded with Icariin (Ica) coated on the stent surface promotes the growth and differentiation of bone cells through the drug-scaffold synergistic effect. Both in vivo and in vitro experiments have shown that the synergistic effect of the composite stent with Icariin significantly enhances the repair of bone defects. This study provides a promising tissue engineering method for the repair of irregular bone defects.
Collapse
Affiliation(s)
- Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuhao Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Weiming Zhao
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China
| | - Zhen Zhang
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liang Qiao
- Henan Univ Sci & Technol, Affiliated Hosp 1, Key Lab Neuromol Biol, Coll Clin Med, Luoyang, Henan 471003, China
| | - Haoming Wu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China
| | - Qiao Su
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lanyu Che
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China
| | - Kai Zhou
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Kainan Li
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China.
| | - Jian He
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan 471000, China.
| |
Collapse
|
24
|
Wang Y, Lv H, Ren S, Zhang J, Liu X, Chen S, Zhai J, Zhou Y. Biological Functions of Macromolecular Protein Hydrogels in Constructing Osteogenic Microenvironment. ACS Biomater Sci Eng 2024; 10:5513-5536. [PMID: 39173130 DOI: 10.1021/acsbiomaterials.4c00910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Irreversible bone defects resulting from trauma, infection, and degenerative illnesses have emerged as a significant health concern. Structurally and functionally controllable hydrogels made by bone tissue engineering (BTE) have become promising biomaterials. Natural proteins are able to establish connections with autologous proteins through unique biologically active regions. Hydrogels based on proteins can simulate the bone microenvironment and regulate the biological behavior of stem cells in the tissue niche, making them candidates for research related to bone regeneration. This article reviews the biological functions of various natural macromolecular proteins (such as collagen, gelatin, fibrin, and silk fibroin) and highlights their special advantages as hydrogels. Then the latest research trends on cross-linking modified macromolecular protein hydrogels with improved mechanical properties and composite hydrogels loaded with exogenous micromolecular proteins have been discussed. Finally, the applications of protein hydrogels, such as 3D printed hydrogels, microspheres, and injectable hydrogels, were introduced, aiming to provide a reference for the repair of clinical bone defects.
Collapse
Affiliation(s)
- Yihan Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Huixin Lv
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Sicong Ren
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Jiameng Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Xiuyu Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Sheng Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Jingjie Zhai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| |
Collapse
|
25
|
Chen Y, Luo Y, Hou X, Zhang L, Wang T, Li X, Liu Z, Zhao J, Aierken A, Cai Z, Lu B, Tan S, Zhao X, Chen F, Zhou Z, Zheng L. Natural Affinity Driven Modification by Silicene to Construct a "Thermal Switch" for Tumorous Bone Loss. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404534. [PMID: 39033540 PMCID: PMC11425228 DOI: 10.1002/advs.202404534] [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: 04/29/2024] [Revised: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Tumorous bone defects present significant challenges for surgical bio-reconstruction due to the dual pathological conditions of residual tumor presence and extensive bone loss following excision surgery. To address this challenge, a "thermal switch" smart bone scaffold based on the silicene nanosheet-modified decalcified bone matrix (SNS@DBM) is developed by leveraging the natural affinity between collagen and silicene, which is elucidated by molecular dynamics simulations. Benefitting from its exceptional photothermal ability, biodegradability, and bioactivity, the SNS@DBM "thermal switch" provides an integrated postoperative sequential thermotherapy for tumorous bone loss by exerting three levels of photothermal stimulation (i.e., strong, moderate, and nonstimulation). During the different phases of postoperative bioconstruction, the SNS@DBM scaffold realizes simultaneous residual tumor ablation, tumor recurrence prevention, and bone tissue regeneration. These biological effects are verified in the tumor-bearing nude mice of patient-derived tissue xenografts and critical cranium defect rats. Mechanism research prompts moderate heat stimulus generated by and coordinating with SNSs can upregulate osteogenic genes, promote macrophages M2 polarization, and intensify angiogenesis of H-type vessels. This study introduces a versatile approach to the management of tumorous bone defects.
Collapse
Affiliation(s)
- Yi‐Xing Chen
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Yi‐Ping Luo
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Xiao‐Dong Hou
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Lei Zhang
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Tian‐Long Wang
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Xi‐Fan Li
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Zhi‐Qing Liu
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Jin‐Hui Zhao
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Aihemaitijiang Aierken
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Zhu‐Yun Cai
- Department of OrthopedicsSecond Affiliated Hospital of Naval Medical University415 Fengyang RoadShanghai200003P. R. China
| | - Bing‐Qiang Lu
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Shuo Tan
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Xin‐Yu Zhao
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Feng Chen
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
- Shanghai Key Laboratory of Craniomaxillofacial Development and DiseasesShanghai Stomatological Hospital & School of StomatologyFudan UniversityShanghai201102P. R. China
| | - Zi‐Fei Zhou
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Long‐Po Zheng
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
- Shanghai Tenth People's Hospital Chongming BranchShanghai202150China
| |
Collapse
|
26
|
Shendage SS, Kachare K, Gaikwad K, Naikwade MB, Kashte S, Ghule AV. Bioactive material‑sodium alginate-polyvinyl alcohol composite film scaffold for bone tissue engineering application. Int J Biol Macromol 2024; 276:133860. [PMID: 39009256 DOI: 10.1016/j.ijbiomac.2024.133860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/29/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Road accidents and infection-causing diseases during bone surgery are serious problems in orthopedics, and thus, addressing these pressing challenges is crucial. In the present study, the 70S30C calcium silicate bioactive material (BM) is synthesized by a sustainable approach employing a precipitation method using recycled rice husk and eggshells as a precursor of silica and calcium. Further, 70S30C BM is composited with sodium alginate (SA) and polyvinyl alcohol (PVA), and the films were prepared by solvent casting method. The composite films were prepared without the addition of acid, binder, and crosslinking agents. Further, the films were characterized by BET, XRD, ATR-FTIR, SEM, and EDS mapping. The in vitro bioactivity and biodegradation study is performed in the simulated body fluid (SBF). The in vitro haemolysis study is executed using human blood and the results demonstrate haemocompatibility of the composite films. The ex ovo CAM assay also exhibits good neovascularization. The in vitro and in vivo biocompatibility assay proves its non-toxic nature. Further, the in vivo study reveals that the engineered composite film demonstrates accelerated osteogenesis. This work broadens the orthopedic potential of the composite film and offers bioactivity, haemocompatibility, angiogenesis, non-toxicity, and in vivo osteogenesis which would serve as a potential candidate for bone tissue engineering application.
Collapse
Affiliation(s)
- Shital S Shendage
- Green Nanotechnology Laboratory, Department of Chemistry, Shivaji University, Kolhapur 416004, India
| | - Kranti Kachare
- Green Nanotechnology Laboratory, Department of Chemistry, Shivaji University, Kolhapur 416004, India
| | - Kajal Gaikwad
- Department of Stem Cell and Regenerative Medicine, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Institution Deemed to Be University), Kolhapur, India
| | - Mahesh B Naikwade
- Department of Chemical and Biological Engineering, Gachon University, South Korea
| | - Shivaji Kashte
- Department of Stem Cell and Regenerative Medicine, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Institution Deemed to Be University), Kolhapur, India
| | - Anil Vithal Ghule
- Green Nanotechnology Laboratory, Department of Chemistry, Shivaji University, Kolhapur 416004, India.
| |
Collapse
|
27
|
Wu Y, Liu P, Feng C, Cao Q, Xu X, Liu Y, Li X, Zhu X, Zhang X. 3D printing calcium phosphate ceramics with high osteoinductivity through pore architecture optimization. Acta Biomater 2024; 185:111-125. [PMID: 39002921 DOI: 10.1016/j.actbio.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
The osteoinductivity of 3D printed calcium phosphate (CaP) ceramics has a large gap compared with those prepared by conventional foaming methods, and improving the osteoinductivity of 3D printing CaP ceramics is crucial for successful application in bone regeneration. Pore architecture plays a critical role in osteoinductivity. In this study, CaP ceramics with a hexagonal close-packed (HCP) spherical pore structure were successfully fabricated using DLP printing technology. Additionally, octahedral (Octahedral), diamond (Diamond), and helical (Gyroid) structures were constructed with similar porosity and macropore diameter. CaP ceramics with the HCP structure exhibited higher compression strength (8.39 ± 1.82 MPa) and lower permeability (6.41 × 10-11 m2) compared to the Octahedral, Diamond, and Gyroid structures. In vitro cellular responses indicated that the macropore architecture strongly influenced the local growth rate of osteoblast-formed cell tissue; cells grew uniformly and formed circular rings in the HCP group. Furthermore, the HCP group promoted the expression of osteogenic genes and proteins more effectively than the other three groups. The outstanding osteoinductivity of the HCP group was confirmed in canine intramuscular implantation studies, where the new bone area reached up to 8.02 ± 1.94 % after a 10-week implantation. Additionally, the HCP group showed effective bone regeneration in repairing femoral condyle defects. Therefore, our findings suggest that 3D printed CaP bioceramics with an HCP structure promote osteoinductivity and can be considered as candidates for personalized precise treatment of bone defects in clinical applications. STATEMENT OF SIGNIFICANCE: 1. 3D printing BCP ceramics with high osteoinductivity were constructed through pore architecture optimization. 2. BCP ceramics with HCP structure exhibited relatively higher mechanical strength and lower permeability than those with Octahedral, Diamond and Gyroid structures. 3. BCP ceramics with HCP structure could promote the osteogenic differentiation of MC3T3-E1, and showed the superior in-vivo osteoinductivity and bone regeneration comparing with the other structures.
Collapse
Affiliation(s)
- Yonghao Wu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Puxin Liu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Quanle Cao
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiujuan Xu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Sichuan University, Chengdu 610064, China
| | - Yunyi Liu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Sichuan University, Chengdu 610064, China.
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Sichuan University, Chengdu 610064, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Sichuan University, Chengdu 610064, China
| |
Collapse
|
28
|
Wang X, Wang L, Cheng B, Wan Q, Wang J, Chen J, Zhu Z, Pei X. Mechanochemically Reprogrammed Tantalum Interfaces Enhance Osseointegration Via Immunomodulation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:44451-44466. [PMID: 39141574 DOI: 10.1021/acsami.4c08533] [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: 08/16/2024]
Abstract
Bone and tooth defects can considerably affect the quality of life and health of patients, and orthopedic implants remain the primary method of addressing such defects. However, implant materials cannot coordinate with the immune microenvironment because of their biological inertness, which may lead to implant loosening or failure. Motivated by the microstructure of nacre, we engineered a biomimetic micro/nanoscale topography on a tantalum surface using a straightforward method. This comprised an organized array of tantalum nanotubes arranged in a brick wall structure, with epigallocatechin gallate acting as "mortar." The coating improved the corrosion resistance, biocompatibility, and antioxidant properties. In vitro and in vivo evaluations further confirmed that coatings can create a favorable bone immune microenvironment through the synergistic effects of mechanochemistry and enhance bone integration. This research offers a new viewpoint on the creation of sophisticated functional implants, possessing vast potential for use in the regeneration and repair of bone tissue.
Collapse
Affiliation(s)
- Xu Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Liang Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Bin Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhou Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
29
|
Chen H, Xu J, Dun Z, Yang Y, Wang Y, Shu F, Zhang Z, Liu M. Emulsion electrospun epigallocatechin gallate-loaded silk fibroin/polycaprolactone nanofibrous membranes for enhancing guided bone regeneration. Biomed Mater 2024; 19:055039. [PMID: 39121887 DOI: 10.1088/1748-605x/ad6dc8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Guided bone regeneration (GBR) membranes play an important role in oral bone regeneration. However, enhancing their bone regeneration potential and antibacterial properties is crucial. Herein, silk fibroin (SF)/polycaprolactone (PCL) core-shell nanofibers loaded with epigallocatechin gallate (EGCG) were prepared using emulsion electrospinning. The nanofibrous membranes were characterized via scanning electron microscopy, transmission electron microscopy, Fourier transform infrared spectroscopy, thermogravimetric analysis, water contact angle (CA) measurement, mechanical properties testing, drug release kinetics, and 1,1-diphenyl-2-picryl-hydrazyl radical (DPPH) free radical scavenging assay. Mouse pre-osteoblast MC3T3-E1 cells were used to assess the biological characteristics, cytocompatibility, and osteogenic differentiation potential of the nanofibrous membrane. Additionally, the antibacterial properties againstStaphylococcus aureus (S. aureus)andEscherichia coli (E. coli)were evaluated. The nanofibers prepared by emulsion electrospinning exhibited a stable core-shell structure with a smooth and continuous surface. The tensile strength of the SF/PCL membrane loaded with EGCG was 3.88 ± 0.15 Mpa, the water CA was 50°, and the DPPH clearance rate at 24 h was 81.73% ± 0.07%. The EGCG release rate of membranes prepared by emulsion electrospinning was reduced by 12% within 72 h compared to that of membranes prepared via traditional electrospinning.In vitroexperiments indicate that the core-shell membranes loaded with EGCG demonstrated good cell compatibility and promoted adhesion, proliferation, and osteogenic differentiation of MC3T3-E1 cells. Furthermore, the EGCG-loaded membranes exhibited inhibitory effects onE. coliandS. aureus. These findings indicate that core-shell nanofibrous membranes encapsulated with EGCG prepared using emulsion electrospinning possess good antioxidant, osteogenic, and antibacterial properties, making them potential candidates for research in GBR materials.
Collapse
Affiliation(s)
- Hong Chen
- Department of Prosthodontics, The Affiliated Stomatological Hosptial of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, People's Republic of China
| | - Jiya Xu
- Department of Prosthodontics, The Affiliated Stomatological Hosptial of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, People's Republic of China
| | - Zhiyue Dun
- Department of Prosthodontics, The Affiliated Stomatological Hosptial of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, People's Republic of China
| | - Yi Yang
- Department of Prosthodontics, The Affiliated Stomatological Hosptial of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, People's Republic of China
| | - Yueqiu Wang
- Department of Endodontics, The Affiliated Stomatological Hosptial of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, People's Republic of China
| | - Fei Shu
- Department of Prosthodontics, The Affiliated Stomatological Hosptial of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, People's Republic of China
| | - Zhihao Zhang
- Department of Prosthodontics, The Affiliated Stomatological Hosptial of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, People's Republic of China
| | - Mei Liu
- Department of Prosthodontics, The Affiliated Stomatological Hosptial of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, People's Republic of China
| |
Collapse
|
30
|
Tang X, Wang Y, Liu N, Deng X, Zhou Z, Yu C, Wang Y, Fang K, Wu T. Methacrylated Carboxymethyl Chitosan Scaffold Containing Icariin-Loaded Short Fibers for Antibacterial, Hemostasis, and Bone Regeneration. ACS Biomater Sci Eng 2024; 10:5181-5193. [PMID: 38935742 DOI: 10.1021/acsbiomaterials.4c00707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Bone defects typically result in bone nonunion, delayed or nonhealing, and localized dysfunction, and commonly used clinical treatments (i.e., autologous and allogeneic grafts) have limited results. The multifunctional bone tissue engineering scaffold provides a new treatment for the repair of bone defects. Herein, a three-dimensional porous composite scaffold with stable mechanical support, effective antibacterial and hemostasis properties, and the ability to promote the rapid repair of bone defects was synthesized using methacrylated carboxymethyl chitosan and icariin-loaded poly-l-lactide/gelatin short fibers (M-CMCS-SFs). Icariin-loaded SFs in the M-CMCS scaffold resulted in the sustained release of osteogenic agents, which was beneficial for mechanical reinforcement. Both the porous structure and the use of chitosan facilitate the effective absorption of blood and fluid exudates. Moreover, its superior antibacterial properties could prevent the occurrence of inflammation and infection. When cultured with bone mesenchymal stem cells, the composite scaffold showed a promotion in osteogenic differentiation. Taken together, such a multifunctional composite scaffold showed comprehensive performance in antibacterial, hemostasis, and bone regeneration, thus holding promising potential in the repair of bone defects and related medical treatments.
Collapse
Affiliation(s)
- Xunmeng Tang
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
| | - Yawen Wang
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China
| | - Na Liu
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xinyuan Deng
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
| | - Ziyi Zhou
- Department of Plastic, Reconstructive and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Chenghao Yu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China
| | - Yuanfei Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, China
| | - Kuanjun Fang
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
- Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, State Key Laboratory for Biofibers and Eco-textiles, College of Textiles & Clothing, Qingdao University, Qingdao 266071, China
| | - Tong Wu
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China
| |
Collapse
|
31
|
Chen J, Luo J, Feng J, Wang Y, Lv H, Zhou Y. Spatiotemporal controlled released hydrogels for multi-system regulated bone regeneration. J Control Release 2024; 372:846-861. [PMID: 38955252 DOI: 10.1016/j.jconrel.2024.06.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Bone defect is one of the urgent problems to be solved in clinics, and it is very important to construct efficient scaffold materials to facilitate bone tissue regeneration. Hydrogels, characterized by their unique three-dimensional network structure, serve as excellent biological scaffold materials. Their internal pores are capable of loading osteogenic drugs to expedite bone formation. The rate and quality of new bone formation are intimately linked with immune regulation and vascular remodeling. The strategic sequential release of drugs to balance inflammation and regulate vascular remodeling is crucial for initiating the osteogenic process. Through the design of hydrogel microstructures, it is possible to achieve sequential drug release and the drug action time can be prolonged, thereby catering to the multi-systemic collaborative regulation needs of osteosynthesis. The drug release rate within the hydrogel is governed by swelling control systems, physical control systems, chemical control systems, and environmental control systems. Utilizing these control systems to design hydrogel materials capable of multi-drug delivery optimizes the construction of the bone microenvironment. Consequently, this facilitates the spatiotemporal controlled released of drugs, promoting bone tissue regeneration. This paper reviews the principles of the controlled release system of various sustained-release hydrogels and the advancements in research on hydrogel multi-drug delivery systems for bone tissue regeneration.
Collapse
Affiliation(s)
- Jingxia Chen
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Jiaxin Luo
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Jian Feng
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Yihan Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Huixin Lv
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China.
| |
Collapse
|
32
|
Ganapathy A, Narayanan K, Chen Y, Villani C, George A. Dentin matrix protein 1 and HUVEC-ECM scaffold promote the differentiation of human dental pulp stem cells into endothelial lineage: implications in regenerative medicine. Front Physiol 2024; 15:1429247. [PMID: 39040080 PMCID: PMC11260688 DOI: 10.3389/fphys.2024.1429247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Reprograming of the dental pulp somatic cells to endothelial cells is an attractive strategy for generation of new blood vessels. For tissue regeneration, vascularization of engineered constructs is crucial to improve repair mechanisms. In this study, we show that dentin matrix protein 1 (DMP1) and HUVEC-ECM scaffold enhances the differentiation potential of dental pulp stem cells (DPSCs) to an endothelial phenotype. Our results show that the differentiated DPSCs expressed endothelial markers CD31 and VE-Cadherin (CD144) at 7 and 14 days. Expression of CD31 and VE-Cadherin (CD144) were also confirmed by immunofluorescence. Furthermore, flow cytometry analysis revealed a steady increase in CD31 and VE-Cadherin (CD144) positive cells with DMP1 treatment when compared with control. In addition, integrins specific for endothelial cells were highly expressed during the differentiation process. The endothelial cell signature of differentiated DPSCs were additionally characterized for key endothelial cell markers using gene expression by RT-PCR, Western blotting, immunostaining, and RNA-seq analysis. Furthermore, the angiogenic phenotype was confirmed by tubule and capillary sprout formation. Overall, stimulation of DPSCs by DMP1 and use of HUVEC-ECM scaffold promoted their differentiation into phenotypically, transcriptionally, and functionally differentiated bonafide endothelial cells. This study is novel, physiologically relevant and different from conventional strategies.
Collapse
Affiliation(s)
| | | | | | | | - Anne George
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
33
|
Ribeiro ED, de Santana IHG, Viana MRM, Júnior ESH, Dias JCP, Ferreira-Júnior O, Sant'Ana E. The efficacy of Platelet and Leukocyte Rich Fibrin (L-PRF) in the healing process and bone repair in oral and maxillofacial surgeries: a systematic review. Clin Oral Investig 2024; 28:414. [PMID: 38965076 DOI: 10.1007/s00784-024-05817-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION The search to optimize the healing and bone repair processes in oral and maxillofacial surgeries reflects the constant evolution in clinical practice, driven by the demand for increasingly satisfactory results and the need to minimize postoperative complications. OBJECTIVE To evaluate the efficacy of Platelet and Leukocyte Rich Fibrin (L-PRF) in the healing and bone repair process in oral and maxillofacial surgeries. MATERIALS AND METHODS The systematic review protocol for this study included the definition of the research question, the domain of the study, the databases searched, the search strategy, the inclusion and exclusion criteria, the types of studies to be included, the measures of effect, the methods for screening, data extraction and analysis, and the approach to data synthesis. Systematic literature searches were carried out on Cochrane databases, Web of Science, PubMed, ScienceDirect, Embase and Google Scholar. RESULTS The strategic search in the databases identified 1,159 studies. After removing the duplicates with the Rayyan© software, 946 articles remained. Of these, 30 met the inclusion criteria. After careful evaluation based on the inclusion and exclusion criteria, 8 studies were considered highly relevant and included in the systematic review. CONCLUSION Platelet and Leukocyte Rich Fibrin (L-PRF) has a positive effect on the healing process and bone repair in oral and maxillofacial surgeries.
Collapse
Affiliation(s)
- Eduardo Dias Ribeiro
- Department of Clinical and Social Dentistry (DCOS), Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | | | | | | | | | - Osny Ferreira-Júnior
- Bauru School of Dentistry, University of São Paulo, (FOB-USP), São Paulo, Brazil
| | - Eduardo Sant'Ana
- Bauru School of Dentistry, University of São Paulo, (FOB-USP), São Paulo, Brazil
| |
Collapse
|
34
|
Zhao F, Qiu Y, Liu W, Zhang Y, Liu J, Bian L, Shao L. Biomimetic Hydrogels as the Inductive Endochondral Ossification Template for Promoting Bone Regeneration. Adv Healthc Mater 2024; 13:e2303532. [PMID: 38108565 DOI: 10.1002/adhm.202303532] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Repairing critical size bone defects (CSBD) is a major clinical challenge and requires effective intervention by biomaterial scaffolds. Inspired by the fact that the cartilaginous template-based endochondral ossification (ECO) process is crucial to bone healing and development, developing biomimetic biomaterials to promote ECO is recognized as a promising approach for repairing CSBD. With the unique highly hydrated 3D polymeric network, hydrogels can be designed to closely emulate the physiochemical properties of cartilage matrix to facilitate ECO. In this review, the various preparation methods of hydrogels possessing the specific physiochemical properties required for promoting ECO are introduced. The materiobiological impacts of the physicochemical properties of hydrogels, such as mechanical properties, topographical structures and chemical compositions on ECO, and the associated molecular mechanisms related to the BMP, Wnt, TGF-β, HIF-1α, FGF, and RhoA signaling pathways are further summarized. This review provides a detailed coverage on the materiobiological insights required for the design and preparation of hydrogel-based biomaterials to facilitate bone regeneration.
Collapse
Affiliation(s)
- Fujian Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Yonghao Qiu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Wenjing Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Jia Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, P. R. China
| |
Collapse
|
35
|
Shokri M, Kharaziha M, Ahmadi Tafti H, Dalili F, Mehdinavaz Aghdam R, Baghaban Eslaminejad M. Engineering Wet-Resistant and Osteogenic Nanocomposite Adhesive to Control Bleeding and Infection after Median Sternotomy. Adv Healthc Mater 2024; 13:e2304349. [PMID: 38593272 DOI: 10.1002/adhm.202304349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/13/2024] [Indexed: 04/11/2024]
Abstract
Median sternotomy surgery stands as one of the prevailing strategies in cardiac surgery. In this study, the cutting-edge bone adhesive is designed, inspired by the impressive adhesive properties found in mussels and sandcastle worms. This work has created an osteogenic nanocomposite coacervate adhesive by integrating a cellulose-polyphosphodopamide interpenetrating network, quaternized chitosan, and zinc, gallium-doped hydroxyapatite nanoparticles. This adhesive is characterized by robust catechol-metal coordination which effectively adheres to both hard and soft tissues with a maximum adhesive strength of 900 ± 38 kPa on the sheep sternum bone, surpassing that of commercial bone adhesives. The release of zinc and gallium cations from nanocomposite adhesives and quaternized chitosan matrix imparts remarkable antibacterial properties and promotes rapid blood coagulation, in vitro and ex vivo. It is also proved that this nanocomposite adhesive exhibits significant in vitro bioactivity, stable degradability, biocompatibility, and osteogenic ability. Furthermore, the capacity of nanocomposite coacervate to adhere to bone tissue and support osteogenesis contributes to the successful healing of a sternum bone defect in a rabbit model in vivo. In summary, these nanocomposite coacervate adhesives with promising characteristics are expected to provide solutions to clinical issues faced during median sternotomy surgery.
Collapse
Affiliation(s)
- Mahshid Shokri
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran
- Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | - Hossein Ahmadi Tafti
- Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Dalili
- School of Metallurgy & Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | | | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
36
|
Liu Y, Wang Y, Lin M, Liu H, Pan Y, Wu J, Guo Z, Li J, Yan B, Zhou H, Fan Y, Hu G, Liang H, Zhang S, Siu MFF, Wu Y, Bai J, Liu C. Cellular Scale Curvature in Bioceramic Scaffolds Enhanced Bone Regeneration by Regulating Skeletal Stem Cells and Vascularization. Adv Healthc Mater 2024:e2401667. [PMID: 38923234 DOI: 10.1002/adhm.202401667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Critical-sized segmental bone defects cannot heal spontaneously, leading to disability and significant increase in mortality. However, current treatments utilizing bone grafts face a variety of challenges from donor availability to poor osseointegration. Drugs such as growth factors increase cancer risk and are very costly. Here, a porous bioceramic scaffold that promotes bone regeneration via solely mechanobiological design is reported. Two types of scaffolds with high versus low pore curvatures are created using high-precision 3D printing technology to fabricate pore curvatures radius in the 100s of micrometers. While both are able to support bone formation, the high-curvature pores induce higher ectopic bone formation and increased vessel invasion. Scaffolds with high-curvature pores also promote faster regeneration of critical-sized segmental bone defects by activating mechanosensitive pathways. High-curvature pore recruits skeletal stem cells and type H vessels from both the periosteum and the marrow during the early phase of repair. High-curvature pores have increased survival of transplanted GFP-labeled skeletal stem cells (SSCs) and recruit more host SSCs. Taken together, the bioceramic scaffolds with defined micrometer-scale pore curvatures demonstrate a mechanobiological approach for orthopedic scaffold design.
Collapse
Affiliation(s)
- Yang Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yue Wang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Minmin Lin
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Hongzhi Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yonghao Pan
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Jianqun Wu
- College of Medicine, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Ziyu Guo
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Jiawei Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Bingtong Yan
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Hang Zhou
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yuanhao Fan
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Ganqing Hu
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Haowen Liang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Shibo Zhang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Ming-Fung Francis Siu
- Department of Building and Real Estate, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, 999077, China
| | - Yongbo Wu
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Jiaming Bai
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Chao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| |
Collapse
|
37
|
Chen K, Ha S, Xu L, Liu C, Liu Y, Wu X, Li Z, Wu S, Yang B, Chen Z. Fluorinated hydroxyapatite conditions a favorable osteo-immune microenvironment via triggering metabolic shift from glycolysis to oxidative phosphorylation. J Transl Med 2024; 22:437. [PMID: 38720345 PMCID: PMC11077739 DOI: 10.1186/s12967-024-05261-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Biological-derived hydroxyapatite is widely used as a bone substitute for addressing bone defects, but its limited osteoconductive properties necessitate further improvement. The osteo-immunomodulatory properties hold crucial promise in maintaining bone homeostasis, and precise modulation of macrophage polarization is essential in this process. Metabolism serves as a guiding force for immunity, and fluoride modification represents a promising strategy for modulating the osteoimmunological environment by regulating immunometabolism. In this context, we synthesized fluorinated porcine hydroxyapatite (FPHA), and has demonstrated its enhanced biological properties and osteogenic capacity. However, it remains unknown whether and how FPHA affects the immune microenvironment of the bone defects. METHODS FPHA was synthesized and its composition and structural properties were confirmed. Macrophages were cultured with FPHA extract to investigate the effects of FPHA on their polarization and the related osteo-immune microenvironment. Furthermore, total RNA of these macrophages was extracted, and RNA-seq analysis was performed to explore the underlying mechanisms associated with the observed changes in macrophages. The metabolic states were evaluated with a Seahorse analyzer. Additionally, immunohistochemical staining was performed to evaluate the macrophages response after implantation of the novel bone substitutes in critical size calvarial defects in SD rats. RESULTS The incorporation of fluoride ions in FPHA was validated. FPHA promoted macrophage proliferation and enhanced the expression of M2 markers while suppressing the expression of M1 markers. Additionally, FPHA inhibited the expression of inflammatory factors and upregulated the expression of osteogenic factors, thereby enhancing the osteogenic differentiation capacity of the rBMSCs. RNA-seq analysis suggested that the polarization-regulating function of FPHA may be related to changes in cellular metabolism. Further experiments confirmed that FPHA enhanced mitochondrial function and promoted the metabolic shift of macrophages from glycolysis to oxidative phosphorylation. Moreover, in vivo experiments validated the above results in the calvarial defect model in SD rats. CONCLUSION In summary, our study reveals that FPHA induces a metabolic shift in macrophages from glycolysis to oxidative phosphorylation. This shift leads to an increased tendency toward M2 polarization in macrophages, consequently creating a favorable osteo-immune microenvironment. These findings provide valuable insights into the impact of incorporating an appropriate concentration of fluoride on immunometabolism and macrophage mitochondrial function, which have important implications for the development of fluoride-modified immunometabolism-based bone regenerative biomaterials and the clinical application of FPHA or other fluoride-containing materials.
Collapse
Affiliation(s)
- Kaidi Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Seongmin Ha
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Leyao Xu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chengwu Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yuanxiang Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiayi Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhipeng Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Shiyu Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Bo Yang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Zhuofan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
38
|
Gresham RC, Filler AC, Fok SW, Czachor M, Schmier N, Pearson C, Bahney C, Leach JK. Compliant substrates mitigate the senescence associated phenotype of stress induced mesenchymal stromal cells. J Biomed Mater Res A 2024; 112:770-780. [PMID: 38095311 PMCID: PMC10948313 DOI: 10.1002/jbm.a.37657] [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: 07/12/2023] [Revised: 11/02/2023] [Accepted: 12/02/2023] [Indexed: 12/27/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a promising cell population for musculoskeletal cell-based therapies due to their multipotent differentiation capacity and complex secretome. Cells from younger donors are mechanosensitive, evidenced by changes in cell morphology, adhesivity, and differentiation as a function of substrate stiffness in both two- and three-dimensional culture. However, MSCs from older individuals exhibit reduced differentiation potential and increased senescence, limiting their potential for autologous use. While substrate stiffness is known to modulate cell phenotype, the influence of the mechanical environment on senescent MSCs is poorly described. To address this question, we cultured irradiation induced premature senescent MSCs on polyacrylamide hydrogels and assessed expression of senescent markers, cell morphology, and secretion of inflammatory cytokines. Compared to cells on tissue culture plastic, senescent MSCs exhibited decreased markers of the senescence associated secretory phenotype (SASP) when cultured on 50 kPa gels, yet common markers of senescence (e.g., p21, CDKN2A, CDKN1A) were unaffected. These effects were muted in a physiologically relevant heterotypic mix of healthy and senescent MSCs. Conditioned media from senescent MSCs on compliant substrates increased osteoblast mineralization compared to conditioned media from cells on TCP. Mixed populations of senescent and healthy cells induced similar levels of osteoblast mineralization compared to healthy MSCs, further indicating an attenuation of the senescent phenotype in heterotypic populations. These data indicate that senescent MSCs exhibit a decrease in senescent phenotype when cultured on compliant substrates, which may be leveraged to improve autologous cell therapies for older donors.
Collapse
Affiliation(s)
- Robert C.H. Gresham
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA
| | - Andrea C. Filler
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA
| | - Shierly W. Fok
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA
| | - Molly Czachor
- Steadman Phillippon Research Institute, Vail, CO, USA
| | - Natalie Schmier
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA
| | - Claire Pearson
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA
| | | | - J. Kent Leach
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA
- Department of Biomedical Engineering, UC Davis, Davis, CA, USA
| |
Collapse
|
39
|
Zhao X, Zhuang Y, Cao Y, Cai F, Lv Y, Zheng Y, Yang J, Shi X. Electrospun Biomimetic Periosteum Capable of Controlled Release of Multiple Agents for Programmed Promoting Bone Regeneration. Adv Healthc Mater 2024; 13:e2303134. [PMID: 38348511 DOI: 10.1002/adhm.202303134] [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: 09/18/2023] [Revised: 01/29/2024] [Indexed: 05/08/2024]
Abstract
The effective repair of large bone defects remains a major challenge due to its limited self-healing capacity. Inspired by the structure and function of the natural periosteum, an electrospun biomimetic periosteum is constructed to programmatically promote bone regeneration using natural bone healing mechanisms. The biomimetic periosteum is composed of a bilayer with an asymmetric structure in which an aligned electrospun poly(ε-caprolactone)/gelatin/deferoxamine (PCL/GEL/DFO) layer mimics the outer fibrous layer of the periosteum, while a random coaxial electrospun PCL/GEL/aspirin (ASP) shell and PCL/silicon nanoparticles (SiNPs) core layer mimics the inner cambial layer. The bilayer controls the release of ASP, DFO, and SiNPs to precisely regulate the inflammatory, angiogenic, and osteogenic phases of bone repair. The random coaxial inner layer can effectively antioxidize, promoting cell recruitment, proliferation, differentiation, and mineralization, while the aligned outer layer can promote angiogenesis and prevent fibroblast infiltration. In particular, different stages of bone repair are modulated in a rat skull defect model to achieve faster and better bone regeneration. The proposed biomimetic periosteum is expected to be a promising candidate for bone defect healing.
Collapse
Affiliation(s)
- Xingkai Zhao
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yu Zhuang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yongjian Cao
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Fengying Cai
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yicheng Lv
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yunquan Zheng
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| |
Collapse
|
40
|
Wang S, Jia Z, Dai M, Feng X, Tang C, Liu L, Cao L. Advances in natural and synthetic macromolecules with stem cells and extracellular vesicles for orthopedic disease treatment. Int J Biol Macromol 2024; 268:131874. [PMID: 38692547 DOI: 10.1016/j.ijbiomac.2024.131874] [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: 10/15/2023] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Serious orthopedic disorders resulting from myriad diseases and impairments continue to pose a considerable challenge to contemporary clinical care. Owing to its limited regenerative capacity, achieving complete bone tissue regeneration and complete functional restoration has proven challenging with existing treatments. By virtue of cellular regenerative and paracrine pathways, stem cells are extensively utilized in the restoration and regeneration of bone tissue; however, low survival and retention after transplantation severely limit their therapeutic effect. Meanwhile, biomolecule materials provide a delivery platform that improves stem cell survival, increases retention, and enhances therapeutic efficacy. In this review, we present the basic concepts of stem cells and extracellular vesicles from different sources, emphasizing the importance of using appropriate expansion methods and modification strategies. We then review different types of biomolecule materials, focusing on their design strategies. Moreover, we summarize several forms of biomaterial preparation and application strategies as well as current research on biomacromolecule materials loaded with stem cells and extracellular vesicles. Finally, we present the challenges currently impeding their clinical application for the treatment of orthopedic diseases. The article aims to provide researchers with new insights for subsequent investigations.
Collapse
Affiliation(s)
- Supeng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China; Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China; Ningxia Medical University, Ningxia 750004, China
| | - Zhiqiang Jia
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Xujun Feng
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China
| | - Chengxuan Tang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Lingling Cao
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China.
| |
Collapse
|
41
|
Xia Y, Zhou R, Wang S, Teng L, Zhang X, Guo Z, Xu Y, Liu W. The design of an RGD in situ sustained delivery system utilizing scallop byssal protein through genetic engineering. Int J Biol Macromol 2024; 267:131636. [PMID: 38641287 DOI: 10.1016/j.ijbiomac.2024.131636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/10/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024]
Abstract
Although bioactive peptides enhancing bone healing have demonstrated effectiveness in treating bone defects, in vivo instability poses a challenge to their clinical application. Currently reported peptide delivery systems do not meet the demands of bone tissue repair regarding stability and peptide release efficacy. Herein, the self-assembling recombinant chimeric protein (Sbp5-2RGD) is developed by genetic engineering with cell adhesion peptide RGD as the targeted peptide and a newly discovered scallop byssal-derived protein Sbp5-2 that can assemble into wet stable films as the structural domain. In vitro studies show that the Sbp5-2RGD film exhibits excellent extensibility and biocompatibility. In vitro and in vivo degradation experiments demonstrate that the film remains stable due to the layer-by-layer degradation mode, resulting in sustained delivery of RGD in situ for up to 4 weeks. Consequently, the film can effectively promote osteogenesis, which accelerates bone defect healing and the implants osseointegration. Cell-level studies further show that the film up-regulates the expression of genes and proteins (ALP, OCN, OSX, OPN, RUNX2, VEGF) associated with osteogenesis and angiogenesis. Overall, this novel protein film represents an intelligent platform for peptide immobilization, protection, and release through its self-assembly, dense structure, and degradation mode, providing a therapeutic strategy for bone repair.
Collapse
Affiliation(s)
- Yinhuan Xia
- Fang Zongxi Center, MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266071, China
| | - Rong Zhou
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuang Wang
- Fang Zongxi Center, MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Luyao Teng
- Fang Zongxi Center, MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Xiaokang Zhang
- Fang Zongxi Center, MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Zhen Guo
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Weizhi Liu
- Fang Zongxi Center, MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266071, China.
| |
Collapse
|
42
|
Ma L, Dong W, Lai E, Wang J. Silk fibroin-based scaffolds for tissue engineering. Front Bioeng Biotechnol 2024; 12:1381838. [PMID: 38737541 PMCID: PMC11084674 DOI: 10.3389/fbioe.2024.1381838] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Silk fibroin is an important natural fibrous protein with excellent prospects for tissue engineering applications. With profound studies in recent years, its potential in tissue repair has been developed. A growing body of literature has investigated various fabricating methods of silk fibroin and their application in tissue repair. The purpose of this paper is to trace the latest developments of SF-based scaffolds for tissue engineering. In this review, we first presented the primary and secondary structures of silk fibroin. The processing methods of SF scaffolds were then summarized. Lastly, we examined the contribution of new studies applying SF as scaffolds in tissue regeneration applications. Overall, this review showed the latest progress in the fabrication and utilization of silk fibroin-based scaffolds.
Collapse
Affiliation(s)
- Li Ma
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
| | - Wenyuan Dong
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
| | - Enping Lai
- College of Biological and Chemical Engineering, Guangxi University of Science and Technology, Liuzhou, China
| | - Jiamian Wang
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
| |
Collapse
|
43
|
Shen HY, Xing F, Shang SY, Jiang K, Kuzmanović M, Huang FW, Liu Y, Luo E, Edeleva M, Cardon L, Huang S, Xiang Z, Xu JZ, Li ZM. Biomimetic Mineralized 3D-Printed Polycaprolactone Scaffold Induced by Self-Adaptive Nanotopology to Accelerate Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:18658-18670. [PMID: 38587811 DOI: 10.1021/acsami.4c02636] [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: 04/09/2024]
Abstract
Three-dimensional (3D)-printed biodegradable polymer scaffolds are at the forefront of personalized constructs for bone tissue engineering. However, it remains challenging to create a biological microenvironment for bone growth. Herein, we developed a novel yet feasible approach to facilitate biomimetic mineralization via self-adaptive nanotopography, which overcomes difficulties in the surface biofunctionalization of 3D-printed polycaprolactone (PCL) scaffolds. The building blocks of self-adaptive nanotopography were PCL lamellae that formed on the 3D-printed PCL scaffold via surface-directed epitaxial crystallization and acted as a linker to nucleate and generate hydroxyapatite crystals. Accordingly, a uniform and robust mineralized layer was immobilized throughout the scaffolds, which strongly bound to the strands and had no effect on the mechanical properties of the scaffolds. In vitro cell culture experiments revealed that the resulting scaffold was biocompatible and enhanced the proliferation and osteogenic differentiation of mouse embryolous osteoblast cells. Furthermore, we demonstrated that the resulting scaffold showed a strong capability to accelerate in vivo bone regeneration using a rabbit bone defect model. This study provides valuable opportunities to enhance the application of 3D-printed scaffolds in bone repair, paving the way for translation to other orthopedic implants.
Collapse
Affiliation(s)
- Hui-Yuan Shen
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Fei Xing
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Si-Yuan Shang
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Kai Jiang
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Maja Kuzmanović
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Fu-Wen Huang
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mariya Edeleva
- Centre for Polymer and Material Technologies, Department of Materials, Textiles and Chemical Engineering, Ghent University, Ghent 9052, Belgium
| | - Ludwig Cardon
- Centre for Polymer and Material Technologies, Department of Materials, Textiles and Chemical Engineering, Ghent University, Ghent 9052, Belgium
| | - Shishu Huang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhou Xiang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhong-Ming Li
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
44
|
Xu S, Tian G, Zhi M, Liu Z, Du Y, Lu X, Li M, Bai J, Li X, Deng J, Ma S, Wang Y. Functionalized PLGA Microsphere Loaded with Fusion Peptide for Therapy of Bone Defects. ACS Biomater Sci Eng 2024; 10:2463-2476. [PMID: 38445948 DOI: 10.1021/acsbiomaterials.3c01858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
The challenges in the treatment of extensive bone defects are infection control and bone regeneration. Bone tissue engineering is currently one of the most promising strategies. In this study, a short biopeptide with specific osteogenic ability is designed by fusion peptide technology and encapsulated with chitosan-modified poly(lactic acid-glycolic acid) (PLGA) microspheres. The fusion peptide (FP) mainly consists of an osteogenic functional sequence (P-15) and a bone-specific binding sequence (Asp-6), which can regulate bone formation accurately and efficiently. Chitosan-modified PLGA with antimicrobial and pro-healing effects is used to achieve the sustained release of fusion peptides. In the early stage, the antimicrobial and soft tissue healing effects can stop the wound infection as soon as possible, which is relevant for the subsequent bone regeneration process. Our data show that CS-PLGA@FP microspheres have antibacterial and pro-cell migration effects in vitro and excellent pro-wound-healing effects in vivo. In addition, CS-PLGA@FP microspheres promote the expression of osteogenic-related factors and show excellent bone regeneration in a rat defect model. Therefore, CS-PLGA@FP microspheres are an efficient biomaterial that can accelerate the recovery of bone defects.
Collapse
Affiliation(s)
- Shendan Xu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Guangjie Tian
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Min Zhi
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Zihao Liu
- Zhongnuo Dental Hospital, Tianjin Nankai District, Tianjin 300101, China
| | - Yaqi Du
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Xuemei Lu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Minting Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Jin Bai
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Xuewen Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Jiayin Deng
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Shiqing Ma
- Department of Stomatology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yonglan Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| |
Collapse
|
45
|
Yang J, Tan Q, Li K, Liao J, Hao Y, Chen Y. Advances and Trends of Photoresponsive Hydrogels for Bone Tissue Engineering. ACS Biomater Sci Eng 2024; 10:1921-1945. [PMID: 38457377 DOI: 10.1021/acsbiomaterials.3c01485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
The development of static hydrogels as an optimal choice for bone tissue engineering (BTE) remains a difficult challenge primarily due to the intricate nature of bone healing processes, continuous physiological functions, and pathological changes. Hence, there is an urgent need to exploit smart hydrogels with programmable properties that can effectively enhance bone regeneration. Increasing evidence suggests that photoresponsive hydrogels are promising bioscaffolds for BTE due to their advantages such as controlled drug release, cell fate modulation, and the photothermal effect. Here, we review the current advances in photoresponsive hydrogels. The mechanism of photoresponsiveness and its advanced applications in bone repair are also elucidated. Future research would focus on the development of more efficient, safer, and smarter photoresponsive hydrogels for BTE. This review is aimed at offering comprehensive guidance on the trends of photoresponsive hydrogels and shedding light on their potential clinical application in BTE.
Collapse
Affiliation(s)
- Juan Yang
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu 610041, PR China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Qingqing Tan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Ka Li
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Ying Hao
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yuwen Chen
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu 610041, PR China
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu 610041, PR China
| |
Collapse
|
46
|
Abbadessa A, Ronca A, Salerno A. Integrating bioprinting, cell therapies and drug delivery towards in vivo regeneration of cartilage, bone and osteochondral tissue. Drug Deliv Transl Res 2024; 14:858-894. [PMID: 37882983 DOI: 10.1007/s13346-023-01437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
The biological and biomechanical functions of cartilage, bone and osteochondral tissue are naturally orchestrated by a complex crosstalk between zonally dependent cells and extracellular matrix components. In fact, this crosstalk involves biomechanical signals and the release of biochemical cues that direct cell fate and regulate tissue morphogenesis and remodelling in vivo. Three-dimensional bioprinting introduced a paradigm shift in tissue engineering and regenerative medicine, since it allows to mimic native tissue anisotropy introducing compositional and architectural gradients. Moreover, the growing synergy between bioprinting and drug delivery may enable to replicate cell/extracellular matrix reciprocity and dynamics by the careful control of the spatial and temporal patterning of bioactive cues. Although significant advances have been made in this direction, unmet challenges and open research questions persist. These include, among others, the optimization of scaffold zonality and architectural features; the preservation of the bioactivity of loaded active molecules, as well as their spatio-temporal release; the in vitro scaffold maturation prior to implantation; the pros and cons of each animal model and the graft-defect mismatch; and the in vivo non-invasive monitoring of new tissue formation. This work critically reviews these aspects and reveals the state of the art of using three-dimensional bioprinting, and its synergy with drug delivery technologies, to pattern the distribution of cells and/or active molecules in cartilage, bone and osteochondral engineered tissues. Most notably, this work focuses on approaches, technologies and biomaterials that are currently under in vivo investigations, as these give important insights on scaffold performance at the implantation site and its interaction/integration with surrounding tissues.
Collapse
Affiliation(s)
- Anna Abbadessa
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), IDIS Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain.
| | - Alfredo Ronca
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125, Naples, Italy.
| | - Aurelio Salerno
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80125, Naples, Italy.
| |
Collapse
|
47
|
Wang K, Zhou M, Zhang Y, Jin Y, Xue Y, Mao D, Rui Y. Fibromodulin facilitates the osteogenic effect of Masquelet's induced membrane by inhibiting the TGF-β/SMAD signaling pathway. Biomater Sci 2024; 12:1898-1913. [PMID: 38426394 DOI: 10.1039/d3bm01665j] [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: 03/02/2024]
Abstract
Masquelet's induced membrane (IM) technique is a promising treatment strategy for the repair of substantial bone defects. The formation of an IM around polymethylmethacrylate bone cement plays a crucial role in this technique. Several studies have indicated that IMs have bioactivity because they contain abundant blood vessels, a variety of cells, and biological factors. The bioactivity of an IM increases during the initial stages of formation, thereby facilitating bone regeneration and remodeling. Nevertheless, the precise mechanisms underlying the enhancement of IM bioactivity and the promotion of bone regeneration necessitate further investigation. In this study, we successfully developed a Masquelet IM model of critical femur defects in rats. By employing proteomics analysis and biological detection techniques, we identified fibromodulin (FMOD) as a pivotal factor contributing to angiogenesis and the enhanced bioactivity of the IM. A significant increase in angiogenesis and the expression of bioactive factors in the IM was also observed with the upregulation of FMOD expression. Furthermore, this effect is mediated through the inhibition of the transforming growth factor beta (TGF-β)/SMAD signaling pathway. We also demonstrated that administering recombinant human FMOD enhanced osteogenesis in rat bone marrow mesenchymal stem cells and angiogenesis in human umbilical vein endothelial cells in vitro. Furthermore, the negative regulatory effect of the TGF-β signaling pathway was verified. In conclusion, this study provides a novel theoretical basis for the application of IMs in bone-defect reconstruction and explores possible new mechanisms that may play an important role in promoting the bioactivity and osteogenic potential of IMs.
Collapse
Affiliation(s)
- Kai Wang
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214062, China.
- Suzhou Medical College of Soochow University, Suzhou, 215031, China
| | - Ming Zhou
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214062, China.
- Suzhou Medical College of Soochow University, Suzhou, 215031, China
| | - Yuanshu Zhang
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214062, China.
| | - Yesheng Jin
- Suzhou Medical College of Soochow University, Suzhou, 215031, China
| | - Yuan Xue
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214062, China.
| | - Dong Mao
- Orthopaedic Institute, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214062, China
| | - Yongjun Rui
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214062, China.
| |
Collapse
|
48
|
Kwon H, Lee S, Byun H, Huh SJ, Lee E, Kim E, Lee J, Shin H. Engineering pre-vascularized 3D tissue and rapid vascular integration with host blood vessels via co-cultured spheroids-laden hydrogel. Biofabrication 2024; 16:025029. [PMID: 38447223 DOI: 10.1088/1758-5090/ad30c6] [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: 12/28/2023] [Accepted: 03/06/2024] [Indexed: 03/08/2024]
Abstract
Recent advances in regenerative medicine and tissue engineering have enabled the biofabrication of three-dimensional (3D) tissue analogues with the potential for use in transplants and disease modeling. However, the practical use of these biomimetic tissues has been hindered by the challenge posed by reconstructing anatomical-scale micro-vasculature tissues. In this study, we suggest that co-cultured spheroids within hydrogels hold promise for regenerating highly vascularized and innervated tissues, bothin vitroandin vivo. Human adipose-derived stem cells (hADSCs) and human umbilical vein cells (HUVECs) were prepared as spheroids, which were encapsulated in gelatin methacryloyl hydrogels to fabricate a 3D pre-vascularized tissue. The vasculogenic responses, extracellular matrix production, and remodeling depending on parameters like co-culture ratio, hydrogel strength, and pre-vascularization time forin vivointegration with native vessels were then delicately characterized. The co-cultured spheroids with 3:1 ratio (hADSCs/HUVECs) within the hydrogel and with a pliable storage modulus showed the greatest vasculogenic potential, and ultimately formedin vitroarteriole-scale vasculature with a longitudinal lumen structure and a complex vascular network after long-term culturing. Importantly, the pre-vascularized tissue also showed anastomotic vascular integration with host blood vessels after transplantation, and successful vascularization that was positive for both CD31 and alpha-smooth muscle actin covering 18.6 ± 3.6μm2of the luminal area. The described co-cultured spheroids-laden hydrogel can therefore serve as effective platform for engineering 3D vascularized complex tissues.
Collapse
Affiliation(s)
- Hyunseok Kwon
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Sangmin Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Hayeon Byun
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Seung Jae Huh
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Eunjin Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Eunhyung Kim
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Jinkyu Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul 04763, Republic of Korea
| |
Collapse
|
49
|
Lian M, Qiao Z, Qiao S, Zhang X, Lin J, Xu R, Zhu N, Tang T, Huang Z, Jiang W, Shi J, Hao Y, Lai H, Dai K. Nerve Growth Factor-Preconditioned Mesenchymal Stem Cell-Derived Exosome-Functionalized 3D-Printed Hierarchical Porous Scaffolds with Neuro-Promotive Properties for Enhancing Innervated Bone Regeneration. ACS NANO 2024; 18:7504-7520. [PMID: 38412232 DOI: 10.1021/acsnano.3c11890] [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: 02/29/2024]
Abstract
The essential role of the neural network in enhancing bone regeneration has often been overlooked in biomaterial design, leading to delayed or compromised bone healing. Engineered mesenchymal stem cells (MSCs)-derived exosomes are becoming increasingly recognized as potent cell-free agents for manipulating cellular behavior and improving therapeutic effectiveness. Herein, MSCs are stimulated with nerve growth factor (NGF) to regulate exosomal cargoes to improve neuro-promotive potential and facilitate innervated bone regeneration. In vitro cell experiments showed that the NGF-stimulated MSCs-derived exosomes (N-Exos) obviously improved the cellular function and neurotrophic effects of the neural cells, and consequently, the osteogenic potential of the osteo-reparative cells. Bioinformatic analysis by miRNA sequencing and pathway enrichment revealed that the beneficial effects of N-Exos may partly be ascribed to the NGF-elicited multicomponent exosomal miRNAs and the subsequent regulation and activation of the MAPK and PI3K-Akt signaling pathways. On this basis, N-Exos were delivered on the micropores of the 3D-printed hierarchical porous scaffold to accomplish the sustained release profile and extended bioavailability. In a rat model with a distal femoral defect, the N-Exos-functionalized hierarchical porous scaffold significantly induced neurovascular structure formation and innervated bone regeneration. This study provided a feasible strategy to modulate the functional cargoes of MSCs-derived exosomes to acquire desirable neuro-promotive and osteogenic potential. Furthermore, the developed N-Exos-functionalized hierarchical porous scaffold may represent a promising neurovascular-promotive bone reparative scaffold for clinical translation.
Collapse
Affiliation(s)
- Meifei Lian
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Zhiguang Qiao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Shichong Qiao
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Xing Zhang
- State Key Laboratory of Mechanical Systems and Vibration, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jieming Lin
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Ruida Xu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Naifeng Zhu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Tianhong Tang
- Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhuoli Huang
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Wenbo Jiang
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Junyu Shi
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Yongqiang Hao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Hongchang Lai
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Kerong Dai
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
50
|
Wang B, Ye X, Chen G, Zhang Y, Zeng Z, Liu C, Tan Z, Jie X. Fabrication and properties of PLA/β-TCP scaffolds using liquid crystal display (LCD) photocuring 3D printing for bone tissue engineering. Front Bioeng Biotechnol 2024; 12:1273541. [PMID: 38440328 PMCID: PMC10910430 DOI: 10.3389/fbioe.2024.1273541] [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: 08/06/2023] [Accepted: 01/08/2024] [Indexed: 03/06/2024] Open
Abstract
Introduction: Bone defects remain a thorny challenge that clinicians have to face. At present, scaffolds prepared by 3D printing are increasingly used in the field of bone tissue repair. Polylactic acid (PLA) has good thermoplasticity, processability, biocompatibility, and biodegradability, but the PLA is brittle and has poor osteogenic performance. Beta-tricalcium phosphate (β-TCP) has good mechanical properties and osteogenic induction properties, which can make up for the drawbacks of PLA. Methods: In this study, photocurable biodegradable polylactic acid (bio-PLA) was utilized as the raw material to prepare PLA/β-TCP slurries with varying β-TCP contents (β-TCP dosage at 0%, 10%, 20%, 30%, 35% of the PLA dosage, respectively). The PLA/β-TCP scaffolds were fabricated using liquid crystal display (LCD) light-curing 3D printing technology. The characterization of the scaffolds was assessed, and the biological activity of the scaffold with the optimal compressive strength was evaluated. The biocompatibility of the scaffold was assessed through CCK-8 assays, hemocompatibility assay and live-dead staining experiments. The osteogenic differentiation capacity of the scaffold on MC3T3-E1 cells was evaluated through alizarin red staining, alkaline phosphatase (ALP) detection, immunofluorescence experiments, and RT-qPCR assays. Results: The prepared scaffold possesses a three-dimensional network structure, and with an increase in the quantity of β-TCP, more β-TCP particles adhere to the scaffold surface. The compressive strength of PLA/β-TCP scaffolds exhibits a trend of initial increase followed by decrease with an increasing amount of β-TCP, reaching a maximum value of 52.1 MPa at a 10% β-TCP content. Degradation rate curve results indicate that with the passage of time, the degradation rate of the scaffold gradually increases, and the pH of the scaffold during degradation shows an alkaline tendency. Additionally, Live/dead staining and blood compatibility experiments suggest that the prepared PLA/β-TCP scaffold demonstrates excellent biocompatibility. CCK-8 experiments indicate that the PLA/β-TCP group promotes cell proliferation, and the prepared PLA/β-TCP scaffold exhibits a significant ability to enhance the osteogenic differentiation of MC3T3-E1 cells in vitro. Discussion: 3D printed LCD photocuring PLA/β-TCP scaffolds could improve surface bioactivity and lead to better osteogenesis, which may provide a unique strategy for developing bioactive implants in orthopedic applications.
Collapse
Affiliation(s)
- Boqun Wang
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, Guangdong, China
- School of Intelligent Manufacturing, Dongguan Polytechnic, Dongguan, Guangdong, China
| | - Xiangling Ye
- Dongguan Hospital, Guangzhou University of Chinese Medicine, Dongguan, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guocai Chen
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yongqiang Zhang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhikui Zeng
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Cansen Liu
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Zhichao Tan
- Dongguan Hospital, Guangzhou University of Chinese Medicine, Dongguan, Guangdong, China
| | - Xiaohua Jie
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|