1
|
Wang L, Jiang S, Zhou J, Gholipourmalekabadi M, Cao Y, Lin K, Zhuang Y, Yuan C. From hard tissues to beyond: Progress and challenges of strontium-containing biomaterials in regenerative medicine applications. Bioact Mater 2025; 49:85-120. [PMID: 40124596 PMCID: PMC11928986 DOI: 10.1016/j.bioactmat.2025.02.039] [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: 10/20/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Tissue engineering and regenerative medicine have emerged as crucial disciplines focused on the development of new tissues and organs to overcome the limitations of traditional treatments for tissue damage caused by accidents, diseases, or aging. Strontium ion (Sr2+) has garnered significant attention for its multifaceted role in promoting regeneration medicine and therapy, especially in bone tissue regeneration. Recently, numerous studies further confirm that Sr2+ also plays a critical in soft tissue regeneration. This review firstly summarizes the influence of Sr2+ on critical biological processes such as osteogenesis, angiogenesis, immune modulation, matrix synthesis, mineralization, and antioxidative defence mechanisms. Then details the classification, properties, advantages, and limitations of Sr-containing biomaterials (SrBMs). Additionally, this review extends to the current applications of SrBMs in regenerative medicine for diverse tissues, including bone, cartilage, skeletal muscle, dental pulp, cardiac tissue, skin, hair follicles, etc. Moreover, the review addresses the challenges associated with current SrBMs and provides insights for their future designing and applications in regenerative medicine.
Collapse
Affiliation(s)
- Liyun Wang
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Shengjie Jiang
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Jialiang Zhou
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Yuan Cao
- Colorado College, 819 N Tejon Street Box 56, Colorado Springs, 80903, Colorado, USA
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Yu Zhuang
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Changyong Yuan
- School of Stomatology, Xuzhou Medical University, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| |
Collapse
|
2
|
Yao MX, Zheng JC, Wang HC, Lv HZ, Zhang YF, Zhang YQ, Shi TL, Zhu YZ, Zhang YZ, Wang XM, Chen W. Application of biphasic mineralized collagen/polycaprolactone scaffolds in the repair of large load-bearing bone defects: A study in a sheep model. J Orthop Translat 2025; 52:138-149. [PMID: 40322042 PMCID: PMC12047585 DOI: 10.1016/j.jot.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 05/08/2025] Open
Abstract
Objective This study aims to evaluate the efficacy of biphasic mineralized collagen/polycaprolactone (bMC/PCL) scaffolds in repairing large load-bearing bone defects, particularly femoral defects, using a sheep model. Methods The bMC/PCL scaffolds were prepared by combining porous mineralized collagen/polycaprolactone (pMC/PCL) with compact mineralized collagen/polycaprolactone (cMC/PCL). The scaffolds were characterized using scanning electron microscopy to observe the microstructure and compression testing to assess mechanical properties. Twenty female sheep were selected to create a 20 mm femoral defect model, divided into a blank group (no material implanted) and an experimental group (bMC/PCL scaffolds implanted), with 10 sheep in each group. Bone healing and lower limb functional recovery were assessed at 1 month, 3 months, and 6 months postoperatively using Lane-Sandhu scores and visual analog scale scores for lameness. Additionally, bone repair progress was analyzed through X-ray, Micro-CT, and histological analyses. Results Compared with the blank group, the bMC/PCL scaffold group showed significant improvement in bone defect repair. At 3 and 6 months postoperatively, X-ray, Micro-CT scans, and histological staining indicated stable scaffold integration and gradual new bone formation. The Lane-Sandhu scores in the experimental group were 3.60 ± 0.548 and 4.00 ± 0.707 at 3 and 6 months, respectively, whereas the blank group experienced plate/screw breakage leading to fixation failure, with scores of 1, indicating better bone healing in the experimental group. The lameness scores in the experimental group were 2.71 ± 0.97 and 1.48 ± 0.86 at 3 and 6 months, respectively, significantly lower than those in the blank group (p < 0.0001 and p = 0.0002). Micro-CT analysis showed that bone volume to tissue volume ratio increased from 28.07 ± 9.22 % to 62.02 ± 11.82 %, bone mineral density increased from 0.392 ± 0.032 g/cm3 to 0.583 ± 0.125 g/cm3, trabecular thickness increased from 0.690 ± 0.224 mm to 1.049 ± 0.089 mm, and trabecular separation decreased from 2.766 ± 1.183 mm to 0.501 ± 0.268 mm at 3 and 6 months postoperatively. Conclusion This study evaluated the efficacy of bMC/PCL scaffolds in repairing large load-bearing bone defects. The bMC/PCL scaffolds demonstrated good bioactivity and mechanical properties, indicating promising clinical application prospects. Future studies should further verify the safety and efficacy of these scaffolds in a wider range of animal models to support their clinical application. Significance statement The bMC/PCL scaffolds offer a promising solution for large femoral bone defects, with potential for clinical use in orthopedic and trauma surgeries. The translational potential of this article The application of bMC/PCL scaffolds in clinical practice is expected to significantly advance the treatment of large bone defects, particularly weight-bearing bone defects. The study shows that bMC/PCL scaffolds have a significant impact on the repair of large weight-bearing bone defects and functional recovery, indicating their potential for application in orthopedics and trauma care. Specifically, the material's supportive role in weight-bearing bones offers new possibilities for its use in the repair of weight-bearing bone defects. Furthermore, the performance of bMC/PCL scaffolds in bone healing makes them an ideal candidate material for treating various bone defects, with broad clinical application prospects. Further clinical trials are necessary to confirm their safety and efficacy in human patients.
Collapse
Affiliation(s)
- Meng-Xuan Yao
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Jing-Chuan Zheng
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Hai-Cheng Wang
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine in Hebei Province, People's Republic of China
| | - Hong-Zhi Lv
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Yi-Fan Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Yu-Qin Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Tai-Long Shi
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Yan-Ze Zhu
- Beijing Allgens Medical Science and Technology Co., Ltd., Beijing, 102629, People's Republic of China
| | - Ying-Ze Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Xiu-Mei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wei Chen
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| |
Collapse
|
3
|
Sun Y, Yao X, Zhang Y, Zhang W, Zhu C, Shen C, Wang Y, Wang X. Zinc Oxide-Copper Sulfide Nanozyme Hydrogels for Bone Defect Repair by Modulating the Bone Immune Microenvironment and Promoting Osteogenesis/Angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40244716 DOI: 10.1021/acsami.4c23069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Bone defects caused by trauma, tumors, or infections pose significant challenges to clinical treatment because of the complex pathological microenvironment they create. Elevated levels of inflammatory factors and reactive oxygen species (ROS) at the defect site disrupt the bone immune microenvironment, impeding bone regeneration. Concurrently, the vascular damage frequently associated with bone defects leads to hypoxia, further complicating therapeutic efforts. Although bone grafting remains a primary clinical approach, its efficacy is limited by these adverse conditions. In this study, a ZnO-CuS/F127 nanozyme hydrogel with multiple enzymatic activities was manufactured for bone defect repair via the modulation of the bone immune microenvironment and the promotion of osteo-/angiogenesis, which was accomplished via the encapsulation of ZnO-CuS nanoflowers synthesized via calcination into the F127 hydrogel matrix. ZnO-CuS bimetallic nanoenzymes exhibit robust catalase (CAT) and superoxide dismutase-like activities, enabling effective scavenging of diverse ROS species in vitro. In cellular assays, ZnO-CuS/F127 protected bone marrow mesenchymal stem cells [bone mesenchymal stem cells (BMSCs)] from ROS-induced cytotoxicity and promoted macrophage polarization toward the anti-inflammatory M2 phenotype, thus modulating the bone immune microenvironment. The ZnO-CuS/F127 hydrogel demonstrated potent proangiogenic and pro-osteogenic effects, attributed to its ability to upregulate the Wnt/β-catenin signaling pathway while inhibiting the NF-κB pathway in BMSCs, as confirmed by RNA sequencing. In vivo, the hydrogel exhibited exceptional hemostatic performance and facilitated bone defect repair in mouse hemorrhage and rat bone defect models while maintaining high biocompatibility and low cytotoxicity. This study highlights the use of the ZnO-CuS/F127 nanozyme hydrogel as a promising therapeutic strategy for bone defect repair. By modulating the immune microenvironment and promoting angiogenesis and osteogenesis, this multifunctional hydrogel offers innovative insights and a potential clinical solution for addressing the multifaceted challenges of bone regeneration.
Collapse
Affiliation(s)
- Yiwei Sun
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, P. R. China
| | - Xiaxi Yao
- School of Chemistry and Materials Engineering, Suzhou Key Laboratory of Functional Ceramic Materials, Changshu Institute of Technology, Changshu 215500, P. R. China
| | - Yiqun Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, P. R. China
| | - Wei Zhang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, P. R. China
| | - Can Zhu
- School of Chemistry and Materials Engineering, Suzhou Key Laboratory of Functional Ceramic Materials, Changshu Institute of Technology, Changshu 215500, P. R. China
| | - Cailiang Shen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Yuanyin Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, P. R. China
| | - Xianwen Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, P. R. China
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, P. R. China
| |
Collapse
|
4
|
Zhang T, Yang J, Lu Y, Wang Y, Wang X, Li Y, Li W, Wang Y. Synergistic Functions of the Janus Fibrous Membrane for Enhanced Bone Repair. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14873-14887. [PMID: 40013909 DOI: 10.1021/acsami.4c18965] [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: 02/28/2025]
Abstract
The treatment of bone defects presents significant challenges in clinical practice. Guided bone regeneration (GBR) strategies offer a new approach, but existing commercial GBR membranes still lack optimal barrier and osteogenic functions. This study presents the design of a Janus fibrous membrane using classic amphiphilic block copolymers and gelatin methacryloyl containing unsaturated double bonds through a gradient electrospinning process. Specifically, by controlling electrospinning parameters, self-assembly of block copolymers, and secondary photo-cross-linking, differential composition, topological structure, and properties between different layers were achieved, thereby realizing synergistic physiological barrier and repair-promoting functions. By leveraging the Janus effect, it effectively blocks the adverse effects of soft tissue cell ingrowth on bone repair while guiding osteogenic cell proliferation and differentiation. Furthermore, the membrane's functionality is optimized by incorporating the antimicrobial component ε-poly-l-lysine and the osteogenic component niobium. In vivo studies demonstrate the membrane's excellent biocompatibility, antibacterial activity, and remarkable bone regeneration potential in both normal and infectious bone defect animal models. The developed Janus fibrous membrane serves as a versatile platform for biomedical applications, offering the vast potential to effectively address the limitations of current GBR membranes in clinical bone defect treatment.
Collapse
Affiliation(s)
- Tingting Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, China
| | - Jinghong Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, China
| | - Yu Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, China
| | - Yanlan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, China
| | - Xiaoshuang Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, China
| | - Yijiao Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, China
| | - Weichang Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, China
| | - Yan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, China
| |
Collapse
|
5
|
Pan Y, Zhao H, Huang W, Liu S, Qi Y, Huang Y. Metal-Protein Hybrid Materials: Unlocking New Frontiers in Biomedical Applications. Adv Healthc Mater 2025; 14:e2404405. [PMID: 39778029 DOI: 10.1002/adhm.202404405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Metal-protein hybrid materials represent a novel class of functional materials that exhibit exceptional physicochemical properties and tunable structures, rendering them remarkable applications in diverse fields, including materials engineering, biocatalysis, biosensing, and biomedicine. The design and development of multifunctional and biocompatible metal-protein hybrid materials have been the subject of extensive research and a key aspiration for practical applications in clinical settings. This review provides a comprehensive analysis of the design strategies, intrinsic properties, and biomedical applications of these hybrid materials, with a specific emphasis on their potential in cancer therapy, drug and vaccine delivery, antibacterial treatments, and tissue regeneration. Through rational design, stable metal-protein hybrid materials can be synthesized using straightforward methods, enabling them with therapeutic, delivery, immunomodulatory, and other desired functionalities. Finally, the review outlines the existing limitations and challenges associated with metal-protein hybrid materials and evaluates their potential for clinical translation, providing insights into their practical implementation within biomedical applications.
Collapse
Affiliation(s)
- Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Han Zhao
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Wenyong Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Siyang Liu
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| |
Collapse
|
6
|
Sun H, Chen Y, Sang X, Liu Q, Yu H, Hu S, Mao Y, Zhang L. Spatiotemporal regulation of the bone immune microenvironment via a 'Zn 2+-quercetin' hierarchical delivery system for bone regeneration. Regen Biomater 2025; 12:rbaf006. [PMID: 40115376 PMCID: PMC11925500 DOI: 10.1093/rb/rbaf006] [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: 10/08/2024] [Revised: 01/04/2025] [Accepted: 01/25/2025] [Indexed: 03/23/2025] Open
Abstract
The immunoregulation of tissue-engineered bone has emerged as a prominent area for bone defect repair. While this field demonstrates considerable potential, effectively managing relevant factors and maintaining a balanced immune microenvironment in practical applications remain substantial challenges that require resolution. In this study, we tested a novel comprehensive hierarchical delivery system based on the requirements of a natural immune microenvironment for inflammatory factors, to optimize local immune responses through precise regulation of drug release. Quercetin (Que)-loaded zeolite imidazolate framework-8 (ZIF-8) nanoparticles were embedded in gelatin methacrylate to create a drug-release system featuring a Zn2+ shell and quercetin core. In vivo and in vitro studies demonstrated that this dual sustained-release hydrogel-ZIF-8 system can produce low concentrations of Zn2+ at an early stage, resulting in a mild anti-inflammatory effect and proliferation of bone marrow mesenchymal stem cells. Moreover, as inflammation advances, the release of quercetin works synergistically with Zn2+ to enhance anti-inflammatory responses, reconfigure the local microenvironment, and mitigate the inflammatory response that adversely impacts bone health by inhibiting the Nuclear Factor-kappa B (NF-κB) signaling pathway, thereby promoting osteogenic differentiation. This system is pioneering for sequential microenvironment regulation based on its diverse anti-inflammatory properties, offering a novel and comprehensive strategy for bone immune regulation in the clinical treatment of bone defects.
Collapse
Affiliation(s)
- Hengliang Sun
- Graduate School, Anhui Medical University, Hefei, Anhui 230032, China
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
- Department of Plastic Surgery, Second People's Hospital of Wuhu City, Wuhu, Anhui 241001, China
| | - Yedan Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
- Anhui Nerve Regeneration Technology and Medical New Materials Engineering Research Center, School of Life Sciences, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Xiaoqin Sang
- Anhui Nerve Regeneration Technology and Medical New Materials Engineering Research Center, School of Life Sciences, Bengbu Medical University, Bengbu, Anhui 233030, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Qingxiang Liu
- Anhui Nerve Regeneration Technology and Medical New Materials Engineering Research Center, School of Life Sciences, Bengbu Medical University, Bengbu, Anhui 233030, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Haoran Yu
- Anhui Nerve Regeneration Technology and Medical New Materials Engineering Research Center, School of Life Sciences, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Shaojun Hu
- Department of Orthopedics, Huaiyuan County People's Hospital, Bengbu, Anhui 233400, China
| | - Yingji Mao
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
- Anhui Nerve Regeneration Technology and Medical New Materials Engineering Research Center, School of Life Sciences, Bengbu Medical University, Bengbu, Anhui 233030, China
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Li Zhang
- Graduate School, Anhui Medical University, Hefei, Anhui 230032, China
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| |
Collapse
|
7
|
Wang Q, Gao Y, Chen Y, Wang X, Pei Q, Zhang T, Wang C, Pan J. Synergistic Enhancement of Antibacterial and Osteo-Immunomodulatory Activities of Titanium Implants via Dual-Responsive Multifunctional Surfaces. Adv Healthc Mater 2025; 14:e2404260. [PMID: 39690750 DOI: 10.1002/adhm.202404260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Bone implant-associated infections and inflammations, primarily caused by bacteria colonization, frequently result in unsuccessful procedures and pose significant health risks to patients. To mitigate these challenges, the development of engineered implants with spatiotemporal regulation capabilities, designed to inhibit bacterial survival and modulate immune responses in the early stage, while promoting bone defect healing in the late stage is proposed. The implants are functionalized with ε-poly-l-lysine-phenylboronic acid (PP) via dynamic boronic ester bonds, which facilitate its release through a reactive oxygen species (ROS) and pH-responsive strategy, thereby establishing an antibacterial microenvironment on and around the implants. Additionally, the dynamic metal coordination interaction facilitates the loading and sustained release of Sr2+ under an acidic environment, providing immunomodulatory and osteogenic effects. The ROS/pH-responsive feature, coupled with the implant-bone tissue integration process, affords precise spatiotemporal regulation of the Ti-TA-Sr-PP implants. This strategy represents a promising approach for the preparation of advanced bone implants.
Collapse
Affiliation(s)
- Qing Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ya Gao
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yanzheng Chen
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Xuan Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qingguo Pei
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Taiyu Zhang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Changping Wang
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jinsong Pan
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
8
|
Qin K, Huang X, Wang S, Liang J, Fan Z. 3D-Printed In Situ Growth of Bilayer MOF Hydrogels for Accelerated Osteochondral Defect Repair. Adv Healthc Mater 2025; 14:e2403840. [PMID: 39552270 DOI: 10.1002/adhm.202403840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/05/2024] [Indexed: 11/19/2024]
Abstract
Repairing osteochondral (OC) defect presents a significant challenge due to the intricate structural requirements and the unpredictable differentiation pathways of bone marrow mesenchymal stem cells (BMSCs). To address this challenge, a novel biomimetic OC hydrogel scaffold is developed that features a structure of soft and hard components. This scaffold incorporates bilayer metal-organic frameworks (MOFs), specifically ZIF-67 in the upper layer and ZIF-8 in the lower layer, achieved through an in situ printing process. This configuration enables the spatial and temporal modulation of BMSC differentiation by controlling the release of Co2⁺ and Zn2⁺. The results demonstrate that the bilayer MOF hydrogels significantly outperform hydrogels that either lack MOFs or contain a single type of MOF in enhancing repair outcomes in rabbit models of knee OC defects. The improved regenerative efficacy is attributed to the distinct chondrogenic and osteogenic differentiation cues provided by the bilayer MOFs, effectively guiding BMSCs toward enhanced tissue regeneration. This customizable biomimetic OC hydrogel scaffold not only opens new avenues for innovative therapeutic strategies but also holds great promise for widespread clinical applications.
Collapse
Affiliation(s)
- Kaiqi Qin
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou, Gansu, 730000, P. R. China
| | - Xinyue Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou, Gansu, 730000, P. R. China
| | - Shengfeng Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou, Gansu, 730000, P. R. China
| | - Jiachen Liang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou, Gansu, 730000, P. R. China
| | - Zengjie Fan
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou, Gansu, 730000, P. R. China
| |
Collapse
|
9
|
Yang W, Zou Q, Wang C, Ren Y, Zhang R, Lin M, Huang Z, Huangfu M, Lin L, Li W, Li X. Enhancing Bone Regeneration and Osteogenic Quality by n-HA Internalized Osteoblasts Synergized with ON Protein: Mechanistic Insights. ACS APPLIED MATERIALS & INTERFACES 2024; 16:68967-68982. [PMID: 39638777 DOI: 10.1021/acsami.4c16045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Bone scaffolds offer hope for oral jawbone repair, yet improving their osteogenic performance remains a clinical challenge. This study investigates a novel approach to enhance early bone formation and osteogenic quality by coloading hydroxyapatite (HA)─internalized osteoblasts (OHA) and osteonectin (ON) onto various scaffolds. Our findings demonstrated that the OHA could effectively facilitate the early bone regeneration by providing rapid calcium and phosphorus ion release via lysosome-mediated HA degradation, while the ON protein helps in ion deposition, cell proliferation, and matrix mineralization. When the PHA (PCL+HA) scaffold was incorporated with both the OHA and ON, the scaffold exhibited superior pro-osteogenic performance, driven by synergistic effects of rapid ion release from the OHA, slow ion release from the PHA, and upregulation of osteogenesis-related genes. The analyses of mechanisms revealed that the OHA activated MAPK and PI3K-Akt pathways, while ON stimulated calcium and Wnt signaling, collectively promoting the osteogenic potential. The strategy presented in this study paves a promising way for the development of advanced bone scaffolds to improve the bone regeneration quality.
Collapse
Affiliation(s)
- Wei Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qin Zou
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| | - Chenxin Wang
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| | - Yuankun Ren
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rui Zhang
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| | - Mingyue Lin
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| | - Zeyu Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengxin Huangfu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lili Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wei Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiyu Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
10
|
Yang H, Huang S, Zhu X, Chen Y, Xu C, Li R, Bu P, Jiang Y, Li C, Yang J, Chen Z, Peng W, Liu L. Scalable fabrication of freely shapable 3D hierarchical Cu-doped hydroxyapatite scaffolds via rapid gelation for enhanced bone repair. Mater Today Bio 2024; 29:101370. [PMID: 39687795 PMCID: PMC11648777 DOI: 10.1016/j.mtbio.2024.101370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Critical-sized bone defects present a formidable challenge in tissue engineering, necessitating innovative approaches that integrate osteogenesis and angiogenesis for effective repair. Inspired by the hierarchical porous structure of natural bone, this study introduces a novel method for the scalable production of ultra-long, copper-doped hydroxyapatite (Cu-HAp) fibers, utilizing the rapid gelation properties of guar gum (GG) under controlled conditions. These fibers serve as foundational units to fabricate three-dimensional porous scaffolds with a biomimetic hierarchical architecture. The scaffolds exhibit a broad pore size distribution (1-500 μm) and abundant nanoporous features, mimicking the native bone extracellular matrix. Physicochemical characterization and in vitro assays demonstrated that the copper doping significantly enhanced osteogenic and angiogenic activities, with optimized concentrations (0.8 % and 1.2 % Cu) facilitating the upregulation of osteogenesis-related genes and proteins, as well as promoting endothelial cell proliferation. In vivo studies further confirmed the scaffolds' efficacy, with the 1.2Cu-HAp group showing a remarkable increase in bone regeneration (bone volume/total volume ratio: 35.7 ± 1.87 %) within the defect site. This research offers a promising strategy for the rapid fabrication of multifunctional scaffolds that not only support bone tissue repair but also actively accelerate the healing process through enhanced vascularization.
Collapse
Affiliation(s)
- Hui Yang
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou 341000, China
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Sirui Huang
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| | - Xinwei Zhu
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Yasi Chen
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou 341000, China
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Chunming Xu
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Ruohan Li
- Third Clinical College, Gannan Medical University, Ganzhou, 341000, China
| | - Pan Bu
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yufan Jiang
- Third Clinical College, Gannan Medical University, Ganzhou, 341000, China
| | - Changwei Li
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Jie Yang
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Zhenyi Chen
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Weijie Peng
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Lin Liu
- Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
11
|
Shao YF, Wang H, Zhu Y, Peng Y, Bai F, Zhang J, Zhang KQ. Hydroxyapatite/Silk Fibroin Composite Scaffold with a Porous Structure and Mechanical Strength Similar to Cancellous Bone by Electric Field-Induced Gel Technology. ACS APPLIED MATERIALS & INTERFACES 2024; 16:60977-60991. [PMID: 39453828 DOI: 10.1021/acsami.4c12470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Repair and regeneration of bone tissue defects is a multidimensional process that has been highly challenging to date. The artificial bone scaffold materials, which play a core role, still face the conflict that a biofriendly porous structure will reduce the mechanical performance and accelerate degradation. Herein, a multistage porous structured hydroxyapatite (HA)/silk fibroin (SF) composite scaffold (e-HA/SF) was successfully constructed by cleverly utilizing electric field-induced gel technology. The results indicated that the prepared e-HA/SF scaffolds possess biomimetic hierarchical porous structures with a suitable porosity similar to that of cancellous bone. The HA nanocrystals were uniformly encapsulated in the three-dimensional space of the composite scaffold, thus endowing the e-HA/SF composite scaffolds with an enhanced mechanical performance. Notably, the maximum compression stress and Young's modulus of e-HA/SF-2 scaffolds can reach 24.66 ± 0.88 and 28.91 ± 3.19 MPa, respectively, which are equivalent to those of cancellous bone. Such mechanical performance enhancement was previously unattainable through conventional freeze-drying strategies. Moreover, the introduction of bioactive nano-HA can trigger the optimal cell response in both static and dynamic cell culture experiments in vitro. The e-HA/SF composite scaffold developed in this study can better balance the conflict between the porous structure and mechanical and degradation properties of porous scaffolds.
Collapse
Affiliation(s)
- Yun-Fei Shao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Hui Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Yiran Zhu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Yu Peng
- College of Advanced Material Engineering, Jiaxing Nanhu University, Jiaxing 314001, P. R. China
| | - Fengjiao Bai
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Jun Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Ke-Qin Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
12
|
Zhao R, Meng X, Pan Z, Li Y, Qian H, Zhu X, Yang X, Zhang X. Advancements in nanohydroxyapatite: synthesis, biomedical applications and composite developments. Regen Biomater 2024; 12:rbae129. [PMID: 39776858 PMCID: PMC11703556 DOI: 10.1093/rb/rbae129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/15/2024] [Accepted: 10/29/2024] [Indexed: 01/11/2025] Open
Abstract
Nanohydroxyapatite (nHA) is distinguished by its exceptional biocompatibility, bioactivity and biodegradability, qualities attributed to its similarity to the mineral component of human bone. This review discusses the synthesis techniques of nHA, highlighting how these methods shape its physicochemical attributes and, in turn, its utility in biomedical applications. The versatility of nHA is further enhanced by doping with biologically significant ions like magnesium or zinc, which can improve its bioactivity and confer therapeutic properties. Notably, nHA-based composites, incorporating metal, polymeric and bioceramic scaffolds, exhibit enhanced osteoconductivity and osteoinductivity. In orthopedic field, nHA and its composites serve effectively as bone graft substitutes, showing exceptional osteointegration and vascularization capabilities. In dentistry, these materials contribute to enamel remineralization, mitigate tooth sensitivity and are employed in surface modification of dental implants. For cancer therapy, nHA composites offer a promising strategy to inhibit tumor growth while sparing healthy tissues. Furthermore, nHA-based composites are emerging as sophisticated platforms with high surface ratio for the delivery of drugs and bioactive substances, gradually releasing therapeutic agents for progressive treatment benefits. Overall, this review delineates the synthesis, modifications and applications of nHA in various biomedical fields, shed light on the future advancements in biomaterials research.
Collapse
Affiliation(s)
- Rui Zhao
- School of Medicine, Department of Inspection, Jiangsu University, Zhenjiang 212013, China
| | - Xiang Meng
- School of Medicine, Department of Inspection, Jiangsu University, Zhenjiang 212013, China
| | - Zixian Pan
- School of Medicine, Department of Inspection, Jiangsu University, Zhenjiang 212013, China
| | - Yongjia Li
- School of Medicine, Department of Inspection, Jiangsu University, Zhenjiang 212013, China
| | - Hui Qian
- School of Medicine, Department of Inspection, Jiangsu University, Zhenjiang 212013, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
13
|
Kim M, Wang X, Li Y, Lin Z, Collins CP, Liu Y, Ahn Y, Tsal HM, Song JW, Duan C, Zhu Y, Sun C, He TC, Luo Y, Reid RR, Ameer GA. Personalized composite scaffolds for accelerated cell- and growth factor-free craniofacial bone regeneration. Bioact Mater 2024; 41:427-439. [PMID: 39188380 PMCID: PMC11345904 DOI: 10.1016/j.bioactmat.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 08/28/2024] Open
Abstract
Approaches to regenerating bone often rely on integrating biomaterials and biological signals in the form of cells or cytokines. However, from a translational point of view, these approaches are challenging due to the sourcing and quality of the biologic, unpredictable immune responses, complex regulatory paths, and high costs. We describe a simple manufacturing process and a material-centric 3D-printed composite scaffold system (CSS) that offers distinct advantages for clinical translation. The CSS comprises a 3D-printed porous polydiolcitrate-hydroxyapatite composite elastomer infused with a polydiolcitrate-graphene oxide hydrogel composite. Using a micro-continuous liquid interface production 3D printer, we fabricate a precise porous ceramic scaffold with 60 wt% hydroxyapatite resembling natural bone. The resulting scaffold integrates with a thermoresponsive hydrogel composite in situ to fit the defect, which is expected to enhance surface contact with surrounding tissue and facilitate biointegration. The antioxidative properties of citrate polymers prevent long-term inflammatory responses. The CSS stimulates osteogenesis in vitro and in vivo. Within 4 weeks in a calvarial critical-sized bone defect model, the CSS accelerated ECM deposition (8-fold) and mineralized osteoid (69-fold) compared to the untreated. Through spatial transcriptomics, we demonstrated the comprehensive biological processes of CSS for prompt osseointegration. Our material-centric approach delivers impressive osteogenic properties and streamlined manufacturing advantages, potentially expediting clinical application for bone reconstruction surgeries.
Collapse
Affiliation(s)
- Mirae Kim
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Xinlong Wang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yiming Li
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Zitong Lin
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Caralyn P. Collins
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208 USA
| | - Yugang Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yujin Ahn
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61820, USA
| | - Hsiu-Ming Tsal
- Department of Radiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Joseph W. Song
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Chongwen Duan
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Cheng Sun
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208 USA
| | - Tong-Chuan He
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Clinical and Translational Sciences Institute, Northwestern University, Chicago, IL, 60611, USA
- Center for Collaborative AI in Healthcare, Institute for AI in Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Russell R. Reid
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Guillermo A. Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Chemistry of Life Process Institute, Northwestern University, Chicago, IL, 60208, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
14
|
Lin CH, Srioudom JR, Sun W, Xing M, Yan S, Yu L, Yang J. The use of hydrogel microspheres as cell and drug delivery carriers for bone, cartilage, and soft tissue regeneration. BIOMATERIALS TRANSLATIONAL 2024; 5:236-256. [PMID: 39734701 PMCID: PMC11681182 DOI: 10.12336/biomatertransl.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 12/31/2024]
Abstract
Bone, cartilage, and soft tissue regeneration is a complex process involving many cellular activities across various cell types. Autografts remain the "gold standard" for the regeneration of these tissues. However, the use of autografts is associated with many disadvantages, including donor scarcity, the requirement of multiple surgeries, and the risk of infection. The development of tissue engineering techniques opens new avenues for enhanced tissue regeneration. Nowadays, the expectations of tissue engineering scaffolds have gone beyond merely providing physical support for cell attachment. Ideal scaffolds should also provide biological cues to actively boost tissue regeneration. As a new type of injectable biomaterial, hydrogel microspheres have been increasingly recognised as promising therapeutic carriers for the local delivery of cells and drugs to enhance tissue regeneration. Compared to traditional tissue engineering scaffolds and bulk hydrogel, hydrogel microspheres possess distinct advantages, including less invasive delivery, larger surface area, higher transparency for visualisation, and greater flexibility for functionalisation. Herein, we review the materials characteristics of hydrogel microspheres and compare their fabrication approaches, including microfluidics, batch emulsion, electrohydrodynamic spraying, lithography, and mechanical fragmentation. Additionally, based on the different requirements for bone, cartilage, nerve, skin, and muscle tissue regeneration, we summarize the applications of hydrogel microspheres as cell and drug delivery carriers for the regeneration of these tissues. Overall, hydrogel microspheres are regarded as effective therapeutic delivery carriers to enhance tissue regeneration in regenerative medicine. However, significant effort is required before hydrogel microspheres become widely accepted as commercial products for clinical use.
Collapse
Affiliation(s)
- Chung-Hsun Lin
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jesse R. Srioudom
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Wei Sun
- Leicester International Institute, Dalian University of Technology, Panjin, Liaoning Province, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Canada
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Le Yu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
- Division of Biological and Biomedical Systems, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Jian Yang
- Biomedical Engineering Program, Westlake University, Hangzhou, Zhejiang Province, China
- Research Centre for Industries of the Future, Westlake University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
15
|
Huang B, Li S, Dai S, Lu X, Wang P, Li X, Zhao Z, Wang Q, Li N, Wen J, Liu Y, Wang X, Man Z, Li W, Liu B. Ti 3C 2T x MXene-Decorated 3D-Printed Ceramic Scaffolds for Enhancing Osteogenesis by Spatiotemporally Orchestrating Inflammatory and Bone Repair Responses. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400229. [PMID: 38973266 PMCID: PMC11425883 DOI: 10.1002/advs.202400229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/10/2024] [Indexed: 07/09/2024]
Abstract
Inflammatory responses play a central role in coordinating biomaterial-mediated tissue regeneration. However, precise modulation of dynamic variations in microenvironmental inflammation post-implantation remains challenging. In this study, the traditional β-tricalcium phosphate-based scaffold is remodeled via ultrathin MXene-Ti3C2 decoration and Zn2+/Sr2+ ion-substitution, endowing the scaffold with excellent reactive oxygen species-scavenging ability, near-infrared responsivity, and enhanced mechanical properties. The induction of mild hyperthermia around the implant via periodic near-infrared irradiation facilitates spatiotemporal regulation of inflammatory cytokines secreted by a spectrum of macrophage phenotypes. The process initially amplifies the pro-inflammatory response, then accelerates M1-to-M2 macrophage polarization transition, yielding a satisfactory pattern of osteo-immunomodulation during the natural bone healing process. Later, sustained release of Zn2+/Sr2+ ions with gradual degradation of the 3D scaffold maintains the favorable reparative M2-dominated immunological microenvironment that supports new bone mineralization. Precise temporal immunoregulation of the bone healing process by the intelligent 3D scaffold enhances bone regeneration in a rat cranial defect model. This strategy paves the way for the application of β-tricalcium phosphate-based materials to guide the dynamic inflammatory and bone tissue responses toward a favorable outcome, making clinical treatment more predictable and durable. The findings also demonstrate that near-infrared irradiation-derived mild hyperthermia is a promising method of immunomodulation.
Collapse
Affiliation(s)
- Benzhao Huang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Shishuo Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
| | - Shimin Dai
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
| | - Xiaoqing Lu
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Peng Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
| | - Xiao Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
| | - Zhibo Zhao
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
| | - Qian Wang
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructure, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210023, P. R. China
| | - Ningbo Li
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, P. R. China
| | - Jie Wen
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, P. R. China
| | - Yifang Liu
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, P. R. China
| | - Xin Wang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, P. R. China
| | - Zhentao Man
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250062, P. R. China
| | - Wei Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Bing Liu
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, P. R. China
| |
Collapse
|
16
|
Wang S, Lei H, Mi Y, Ma P, Fan D. Chitosan and hyaluronic acid based injectable dual network hydrogels - Mediating antimicrobial and inflammatory modulation to promote healing of infected bone defects. Int J Biol Macromol 2024; 274:133124. [PMID: 38897505 DOI: 10.1016/j.ijbiomac.2024.133124] [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: 02/29/2024] [Revised: 05/09/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
In bone defects, infections lead to excessive inflammation, increased bacterial, and bone lysis, resulting in irregular wounds that hinder new bone regeneration. Injectable bioactive materials with adequate antimicrobial activity and strong osteogenic potential are urgently required to remedy irregular defects, eradicate bacteria, and facilitate the generation of new bone tissue. In this research, injectable dual-network composite hydrogels consisting of sulfated chitosan, oxidized hyaluronic acid, β-sodium glycerophosphate, and CuSr doped mesoporous bioactive glass loaded with bone morphogenetic protein (CuSrMBGBMP-2) were utilized for the first time to treat infectious bone defects. Initially, the hydrogel was injected into the wound at 37 °C with minimal invasion to establish a stable state and prevent hydrogel loss. Subsequently, sulfated chitosan eliminated bacteria at the wound site and facilitated cell proliferation with oxidized hyaluronic acid. Additionally, CuSrMBGBMP-2 strengthened antibacterial properties, regulated inflammatory reactions, promoted angiogenesis and osteogenic differentiation, addressing the deficiency in late-stage osteogenesis. Specifically, the injectable dual-network hydrogel based on chitosan and hyaluronic acid is minimally invasive, offering antibacterial, anti-inflammatory, pro-angiogenic, and bone regeneration properties. Therefore, this hydrogel with injectable dual network properties holds great promise for the treatment of bone infections in the future.
Collapse
Affiliation(s)
- Shang Wang
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| | - Huan Lei
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| | - Yu Mi
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| | - Pei Ma
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| |
Collapse
|
17
|
Li Y, Xu C, Mao J, Mao L, Li W, Liu Z, Shin A, Wu J, Hou L, Li D, Lin K, Liu J. ZIF-8-based Nanoparticles for Inflammation Treatment and Oxidative Stress Reduction in Periodontitis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36077-36094. [PMID: 38949426 DOI: 10.1021/acsami.4c05722] [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: 07/02/2024]
Abstract
Periodontitis, an inflammatory bone resorption disease associated with dental plaque, poses significant challenges for effective treatment. In this study, we developed Mino@ZIF-8 nanoparticles inspired by the periodontal microenvironment and the unique properties of zeolitic imidazolate framework 8, aiming to address the complex pathogenesis of periodontitis. Transcriptome analysis revealed the active engagement of Mino@ZIF-8 nanoparticles in innate and adaptive inflammatory host defense and cellular metabolic remodeling. Through sustained release of the anti-inflammatory and antibacterial agent minocycline hydrochloride (Mino) and the generation of Zn2+ with pro-antioxidant effects during degradation, Mino@ZIF-8 nanoparticles synergistically alleviate inflammation and oxidative damage. Notably, our study focuses on the pivotal role of zinc ions in mitochondrial oxidation protection. Under lipopolysaccharide (LPS) stimulation, periodontal ligament cells undergo a metabolic shift from oxidative phosphorylation (OXPHOS) to glycolysis, leading to reduced ATP production and increased reactive oxygen species levels. However, Zn2+ effectively rebalances the glycolysis-OXPHOS imbalance, restoring cellular bioenergetics, mitigating oxidative damage, rescuing impaired mitochondria, and suppressing inflammatory cytokine production through modulation of the AKT/GSK3β/NRF2 pathway. This research not only presents a promising approach for periodontitis treatment but also offers novel therapeutic opportunities for zinc-containing materials, providing valuable insights into the design of biomaterials targeting cellular energy metabolism regulation.
Collapse
Affiliation(s)
- Yaxin Li
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Chenci Xu
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Jing Mao
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Lixia Mao
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Weiqi Li
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Ziyang Liu
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Airi Shin
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Jiaqing Wu
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Lingli Hou
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Dejian Li
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201301, China
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Jiaqiang Liu
- Department of Oral and Cranio-Maxillofacial Surgery, 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, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| |
Collapse
|
18
|
Kersey AL, Singh I, Gaharwar AK. Inorganic ions activate lineage-specific gene regulatory networks. Acta Biomater 2024; 183:371-386. [PMID: 38552761 DOI: 10.1016/j.actbio.2024.03.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 03/06/2024] [Accepted: 03/22/2024] [Indexed: 06/14/2024]
Abstract
Inorganic biomaterials have been shown to direct cellular responses, including cell-cell and cell-matrix interactions. Notably, ions released from these inorganic biomaterials play a vital role in defining cell identity, and promoting tissue-specific functions. However, the effect of inorganic ions on cellular functions have yet to be investigated at the transcriptomic level, representing a critical knowledge gap in the development of next-generation bioactive materials. To address this gap, we investigated the impact of various inorganic ions including silver, copper, titanium, and platinum on human mesenchymal stem cells (hMSCs). Our finding showed that silver and copper induce osteogenic and chondrogenic differentiation respectively, through enrichment of lineage-specific gene expression program. In particular, silver effectively induced Wingless/Integrated (Wnt) and mitogen-activated protein kinase (MAPK) signaling, which are vital for osteogenesis. On the other hand, copper specifically stimulated Transforming growth factor beta (TGFβ) signaling, while suppressing Janus kinase/signal transducers and activators of transcription (JAK-STAT) signaling, thereby promoting chondrogenesis. In contrast, platinum, and tantalum, ions didn't stimulate regenerative responses. Together, our findings highlight the potential of inorganic biomaterials in tissue regeneration strategies, which currently rely largely on growth factors and small molecule therapeutics. STATEMENT OF SIGNIFICANCE: This research emphasizes the critical role of bioactive inorganic ions in controlling lineage-specific gene expression patterns in mesenchymal stem cells, effectively modulating the transcriptome landscape and directing cell fate. The study lays the foundation for a systematic database of biomaterial candidates and their effects on cellular functions, which will ultimately streamline the translation of new biomaterials into clinical applications.
Collapse
Affiliation(s)
- Anna L Kersey
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, United States
| | - Irtisha Singh
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, United States; Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, College Station, TX 77807, United States; Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, United States.
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, United States; Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, United States; Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX 77843, United States; Department of Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, United States.
| |
Collapse
|
19
|
Özcolak B, Erenay B, Odabaş S, Jandt KD, Garipcan B. Effects of bone surface topography and chemistry on macrophage polarization. Sci Rep 2024; 14:12721. [PMID: 38830871 PMCID: PMC11148019 DOI: 10.1038/s41598-024-62484-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024] Open
Abstract
Surface structure plays a crucial role in determining cell behavior on biomaterials, influencing cell adhesion, proliferation, differentiation, as well as immune cells and macrophage polarization. While grooves and ridges stimulate M2 polarization and pits and bumps promote M1 polarization, these structures do not accurately mimic the real bone surface. Consequently, the impact of mimicking bone surface topography on macrophage polarization remains unknown. Understanding the synergistic sequential roles of M1 and M2 macrophages in osteoimmunomodulation is crucial for effective bone tissue engineering. Thus, exploring the impact of bone surface microstructure mimicking biomaterials on macrophage polarization is critical. In this study, we aimed to sequentially activate M1 and M2 macrophages using Poly-L-Lactic acid (PLA) membranes with bone surface topographical features mimicked through the soft lithography technique. To mimic the bone surface topography, a bovine femur was used as a model surface, and the membranes were further modified with collagen type-I and hydroxyapatite to mimic the bone surface microenvironment. To determine the effect of these biomaterials on macrophage polarization, we conducted experimental analysis that contained estimating cytokine release profiles and characterizing cell morphology. Our results demonstrated the potential of the hydroxyapatite-deposited bone surface-mimicked PLA membranes to trigger sequential and synergistic M1 and M2 macrophage polarizations, suggesting their ability to achieve osteoimmunomodulatory macrophage polarization for bone tissue engineering applications. Although further experimental studies are required to completely investigate the osteoimmunomodulatory effects of these biomaterials, our results provide valuable insights into the potential advantages of biomaterials that mimic the complex microenvironment of bone surfaces.
Collapse
Affiliation(s)
- Birgün Özcolak
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Berkay Erenay
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey
| | - Sedat Odabaş
- Biomaterials and Tissue Engineering Laboratory (bteLAB), Department of Chemistry, Faculty of Science, Ankara University, 06560, Ankara, Turkey
- Interdisciplinary Research Unit for Advanced Materials (INTRAM), Ankara University, 06560, Ankara, Turkey
| | - Klaus D Jandt
- Chair of Materials Science (CMS), Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Friedrich Schiller University Jena, Löbdergraben 32, 07743, Jena, Germany
| | - Bora Garipcan
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey.
| |
Collapse
|
20
|
Qiu H, Wang J, Hu H, Song L, Liu Z, Xu Y, Liu S, Zhu X, Wang H, Bao C, Lin H. Preparation of an injectable and photocurable carboxymethyl cellulose/hydroxyapatite composite and its application in cranial regeneration. Carbohydr Polym 2024; 333:121987. [PMID: 38494238 DOI: 10.1016/j.carbpol.2024.121987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/08/2024] [Accepted: 02/24/2024] [Indexed: 03/19/2024]
Abstract
Limited bone regeneration, uncontrollable degradation rate, mismatched defect zone and poor operability have plagued the reconstruction of irregular bone defect by tissue-engineered materials. A combination of biomimetic scaffolds with hydroxyapatite has gained great popularity in promoting bone regeneration. Therefore, we designed an injectable, photocurable and in-situ curing hydrogel by methacrylic anhydride -modified carboxymethyl cellulose (CMC-MA) loading with spherical hydroxyapatite (HA) to highly simulate the natural bony matrix and match any shape of damaged tissue. The prepared carboxymethyl cellulose-methacrylate/ hydroxyapatite(CMC-MA/HA) composite presented good rheological behavior, swelling ratio and mechanical property under light illumination. Meanwhile, this composite hydrogel promoted effectively proliferation, supported adhesion and upregulated the osteogenic-related genes expression of MC3T3-E1 cells in vitro, as well as the activity of the osteogenic critical protein, Integrin α1, β1, Myosin 9, Myosin 10, BMP-2 and Smad 1 in Integrin/BMP-2 signal pathway. Together, the composite hydrogels realized promotion of bone regeneration, deformity improvement, and the enhanced new bone strength in skull defect. It also displayed a good histocompatibility and stability of subcutaneous implantation in vivo. Overall, this study laid the groundwork for future research into developing a novel biomaterial and a minimally invasive therapeutic strategies for reconstructing bone defects and contour deficiencies.
Collapse
Affiliation(s)
- He Qiu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jing Wang
- National Engineering Research Center for Biomaterials, College Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Hong Hu
- National Engineering Research Center for Biomaterials, College Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Lu Song
- National Engineering Research Center for Biomaterials, College Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Zhanhong Liu
- National Engineering Research Center for Biomaterials, College Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Yang Xu
- National Engineering Research Center for Biomaterials, College Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Shuo Liu
- National Engineering Research Center for Biomaterials, College Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Hang Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Hai Lin
- National Engineering Research Center for Biomaterials, College Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| |
Collapse
|
21
|
Ding S, Ren T, Song S, Peng C, Liu C, Wu J, Chang X. Combined application of mesenchymal stem cells and different glucocorticoid dosing alleviates osteoporosis in SLE murine models. Immun Inflamm Dis 2024; 12:e1319. [PMID: 38888448 PMCID: PMC11184931 DOI: 10.1002/iid3.1319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 05/23/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE Bone mesenchymal stem cells (BMSCs) have been tentatively applied in the treatment of glucocorticoid-induced osteoporosis (GIOP) and systemic lupus erythematosus (SLE). However, the effects of BMSCs on osteoporosis within the context of glucocorticoid (GC) application in SLE remain unclear. Our aim was to explore the roles of BMSCs and different doses of GC interventions on osteoporosis in SLE murine models. METHODS MRL/MpJ-Faslpr mice were divided into eight groups with BMSC treatment and different dose of GC intervention. Three-dimensional imaging analysis and hematoxylin and eosin (H&E) staining were performed to observe morphological changes. The concentrations of osteoprotegerin (OPG) and receptor activator of nuclear factor κB ligand (RANKL) in serum were measured by enzyme-linked immunosorbent assay (ELISA). The subpopulation of B cells and T cells in bone marrows and spleens were analyzed by flow cytometry. Serum cytokines and chemokines were assessed using Luminex magnetic bead technology. RESULTS BMSCs ameliorated osteoporosis in murine SLE models by enhancing bone mass, improving bone structure, and promoting bone formation through increased bone mineral content and optimization of trabecular morphology. BMSC and GC treatments reduced the number of B cells in bone marrows, but the effect was not significant in spleens. BMSCs significantly promoted the expression of IL-10 while reducing IL-18. Moreover, BMSCs exert immunomodulatory effects by reducing Th17 expression and rectifying the Th17/Treg imbalance. CONCLUSION BMSCs effectively alleviate osteoporosis induced by SLE itself, as well as osteoporosis resulting from SLE combined with various doses of GC therapy. The therapeutic effects of BMSCs appear to be mediated by their influence on bone marrow B cells, T cell subsets, and associated cytokines. High-dose GC treatment exerts a potent anti-inflammatory effect but may hinder the immunotherapeutic potential of BMSCs. Our research may offer valuable guidance to clinicians regarding the use of BMSC treatment in SLE and provide insights into the judicious use of GCs in clinical practice.
Collapse
Affiliation(s)
- Sisi Ding
- Jiangsu Institute of Clinical ImmunologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Tian Ren
- Department of RheumatologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Saizhe Song
- Jiangsu Institute of Clinical ImmunologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Cheng Peng
- Department of RheumatologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Cuiping Liu
- Jiangsu Institute of Clinical ImmunologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jian Wu
- Department of RheumatologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xin Chang
- Department of RheumatologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
22
|
Chen T, Wu X, Zhang P, Wu W, Dai H, Chen S. Strontium-Doped Hydroxyapatite Coating Improves Osteo/Angiogenesis for Ameliorative Graft-Bone Integration via the Macrophage-Derived Cytokines-Mediated Integrin Signal Pathway. ACS APPLIED MATERIALS & INTERFACES 2024; 16:15687-15700. [PMID: 38511302 DOI: 10.1021/acsami.3c14904] [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: 03/22/2024]
Abstract
Polyethylene terephthalate (PET) artificial ligaments, renowned for their superior mechanical properties, have been extensively adopted in anterior cruciate ligament (ACL) reconstruction surgeries. However, the inherent bio-inertness of PET introduces formidable barriers to graft-bone integration, a critical aspect of rehabilitation. Previous interventions, ranging from surface roughening to chemical modifications, have aimed to address this challenge; however, consistently effective techniques for inducing graft-bone integration remain scarce. Our study employed advanced surface-coating methodologies to introduce strontium-doped hydroxyapatite (SrHA) onto PET ligaments. Detailed scanning electron microscopy (SEM) examinations revealed a uniform and integrative coating of SrHA on PET fibers. Furthermore, spectroscopic analysis confirmed the steady release of strontium ions from the coated surface under physiological conditions. In-depth cellular studies proved that extracellular strontium emanating from SrHA-coated PET (PET@SrHA) ligaments actively steers the M2 macrophage polarization. Additionally, macrophages (Mφs) manifested a heightened secretion of prohealing cytokines when exposed to PET@SrHA. Subsequent investigations showed that these cytokines acted as mediators, activating integrin signaling pathways among macrophages, vascular endothelial cells, and osteoblasts. As a direct consequence, an increased rate of angiogenesis and osteogenic differentiation was observed, vital for graft-bone integration following ACL reconstruction with PET@SrHA ligaments. From a biochemical standpoint, our results pinpoint strontium ions as influential immunomodulators, sculpting the graft-bone interface's immune environment. This insight presents the SrHA-coating technique as a viable therapeutic strategy, holding sound promise for improving angiogenesis and osseointegration outcomes during ACL reconstruction using PET-based grafts.
Collapse
Affiliation(s)
- Tianwu Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaopei Wu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Peng Zhang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wei Wu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
23
|
Ming P, Liu Y, Yu P, Jiang X, Yuan L, Cai S, Rao P, Cai R, Lan X, Tao G, Xiao J. A Biomimetic Se-nHA/PC Composite Microsphere with Synergistic Immunomodulatory and Osteogenic Ability to Activate Bone Regeneration in Periodontitis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305490. [PMID: 37852940 DOI: 10.1002/smll.202305490] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/21/2023] [Indexed: 10/20/2023]
Abstract
Accumulation of reactive oxygen species (ROS) in periodontitis exacerbates the destruction of alveolar bone. Therefore, scavenging ROS to reshape the periodontal microenvironment, alleviate the inflammatory response and promote endogenous stem cell osteogenic differentiation may be an effective strategy for treating bone resorption in periodontitis. In this study, sericin-hydroxyapatite nanoparticles (Se-nHA NPs) are synthesized using a biomimetic mineralization method. Se-nHA NPs and proanthocyanidins (PC) are then encapsulated in sericin/sodium alginate (Se/SA) using an electrostatic injection technique to prepare Se-nHA/PC microspheres. Microspheres are effective in scavenging ROS, inhibiting the polarization of macrophages toward the M1 type, and inducing the polarization of macrophages toward the M2 type. In normal or macrophage-conditioned media, the Se-nHA/PC microspheres effectively promoted the osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs). Furthermore, the Se-nHA/PC microspheres demonstrated anti-inflammatory effects in a periodontitis rat model by scavenging ROS and suppressing pro-inflammatory cytokines. The Se-nHA/PC microspheres are also distinguished by their capacity to decrease alveolar bone loss, reduce osteoclast activity, and boost osteogenic factor expression. Therefore, the biomimetic Se-nHA/PC composite microspheres have efficient ROS-scavenging, anti-inflammatory, and osteogenic abilities and can be used as a multifunctional filling material for inflammatory periodontal tissue regeneration.
Collapse
Affiliation(s)
- Piaoye Ming
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Yunfei Liu
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Peiyang Yu
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Xueyu Jiang
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Linlin Yuan
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Shuyu Cai
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Pengcheng Rao
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Rui Cai
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Gang Tao
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Jingang Xiao
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
24
|
Tan J, Li S, Sun C, Bao G, Liu M, Jing Z, Fu H, Sun Y, Yang Q, Zheng Y, Wang X, Yang H. A Dose-Dependent Spatiotemporal Response of Angiogenesis Elicited by Zn Biodegradation during the Initial Stage of Bone Regeneration. Adv Healthc Mater 2024; 13:e2302305. [PMID: 37843190 DOI: 10.1002/adhm.202302305] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/09/2023] [Indexed: 10/17/2023]
Abstract
Zinc (Zn) plays a crucial role in bone metabolism and imbues biodegradable Zn-based materials with the ability to promote bone regeneration in bone trauma. However, the impact of Zn biodegradation on bone repair, particularly its influence on angiogenesis, remains unexplored. This study reveals that Zn biodegradation induces a consistent dose-dependent spatiotemporal response in angiogenesis,both in vivo and in vitro. In a critical bone defect model, an increase in Zn release intensity from day 3 to 10 post-surgery is observed. By day 10, the CD31-positive area around the Zn implant significantly surpasses that of the Ti implant, indicating enhanced angiogenesis. Furthermore,angiogenesis exhibits a distance-dependent pattern closely mirroring the distribution of Zn signals from the implant. In vitro experiments demonstrate that Zn extraction fosters the proliferation and migration of human umbilical vein endothelial cells and upregulates the key genes associated with tube formation, such as HIF-1α and VEGF-A, peaking at a concentration of 22.5 µM. Additionally, Zn concentrations within the range of 11.25-45 µM promote the polarization of M0-type macrophages toward the M2-type, while inhibiting polarization toward the M1-type. These findings provide essential insights into the biological effects of Zn on bone repair, shedding light on its potential applications.
Collapse
Affiliation(s)
- Junlong Tan
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Shuang Li
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Chaoyang Sun
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Guo Bao
- Department of Reproduction and Physiology, National Research Institute for Family Planning, Beijing, 100081, China
| | - Meijing Liu
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Zehao Jing
- Beijing Key Laboratory of Spinal Disease Research, Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Hanwei Fu
- School of Materials Science and Engineering, Beihang University, 37 Xueyuan Rd, Beijing, China
| | - Yanhua Sun
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co. Ltd., Jinan, 250100, China
| | - Qingmin Yang
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co. Ltd., Jinan, 250100, China
| | - Yufeng Zheng
- Beijing Advanced Innovation Center for Materials Genome Engineering and School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xiaogang Wang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Hongtao Yang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| |
Collapse
|
25
|
Zhao Q, Ni Y, Wei H, Duan Y, Chen J, Xiao Q, Gao J, Yu Y, Cui Y, Ouyang S, Miron RJ, Zhang Y, Wu C. Ion incorporation into bone grafting materials. Periodontol 2000 2024; 94:213-230. [PMID: 37823468 DOI: 10.1111/prd.12533] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023]
Abstract
The use of biomaterials in regenerative medicine has expanded to treat various disorders caused by trauma or disease in orthopedics and dentistry. However, the treatment of large and complex bone defects presents a challenge, leading to a pressing need for optimized biomaterials for bone repair. Recent advances in chemical sciences have enabled the incorporation of therapeutic ions into bone grafts to enhance their performance. These ions, such as strontium (for bone regeneration/osteoporosis), copper (for angiogenesis), boron (for bone growth), iron (for chemotaxis), cobalt (for B12 synthesis), lithium (for osteogenesis/cementogenesis), silver (for antibacterial resistance), and magnesium (for bone and cartilage regeneration), among others (e.g., zinc, sodium, and silica), have been studied extensively. This review aims to provide a comprehensive overview of current knowledge and recent developments in ion incorporation into biomaterials for bone and periodontal tissue repair. It also discusses recently developed biomaterials from a basic design and clinical application perspective. Additionally, the review highlights the importance of precise ion introduction into biomaterials to address existing limitations and challenges in combination therapies. Future prospects and opportunities for the development and optimization of biomaterials for bone tissue engineering are emphasized.
Collapse
Affiliation(s)
- Qin Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yueqi Ni
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Hongjiang Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yiling Duan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Jingqiu Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Qi Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Jie Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yiqian Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yu Cui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Simin Ouyang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
26
|
Mi L, Li F, Xu D, Liu J, Li J, Zhong L, Liu Y, Bai N. Performance of 3D printed porous polyetheretherketone composite scaffolds combined with nano-hydroxyapatite/carbon fiber in bone tissue engineering: a biological evaluation. Front Bioeng Biotechnol 2024; 12:1343294. [PMID: 38333080 PMCID: PMC10850574 DOI: 10.3389/fbioe.2024.1343294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Polyetheretherketone (PEEK) has been one of the most promising materials in bone tissue engineering in recent years, with characteristics such as biosafety, corrosion resistance, and wear resistance. However, the weak bioactivity of PEEK leads to its poor integration with bone tissues, restricting its application in biomedical fields. This research effectively fabricated composite porous scaffolds using a combination of PEEK, nano-hydroxyapatite (nHA), and carbon fiber (CF) by the process of fused deposition molding (FDM). The experimental study aimed to assess the impact of varying concentrations of nHA and CF on the biological performance of scaffolds. The incorporation of 10% CF has been shown to enhance the overall mechanical characteristics of composite PEEK scaffolds, including increased tensile strength and improved mechanical strength. Additionally, the addition of 20% nHA resulted in a significant increase in the surface roughness of the scaffolds. The high hydrophilicity of the PEEK composite scaffolds facilitated the in vitro inoculation of MC3T3-E1 cells. The findings of the study demonstrated that the inclusion of 20% nHA and 10% CF in the scaffolds resulted in improved cell attachment and proliferation compared to other scaffolds. This suggests that the incorporation of 20% nHA and 10% CF positively influenced the properties of the scaffolds, potentially facilitating bone regeneration. In vitro biocompatibility experiments showed that PEEK composite scaffolds have good biosafety. The investigation on osteoblast differentiation revealed that the intensity of calcium nodule staining intensified, along with an increase in the expression of osteoblast transcription factors and alkaline phosphatase activities. These findings suggest that scaffolds containing 20% nHA and 10% CF have favorable properties for bone induction. Hence, the integration of porous PEEK composite scaffolds with nHA and CF presents a promising avenue for the restoration of bone defects using materials in the field of bone tissue engineering.
Collapse
Affiliation(s)
- Lian Mi
- Department of Oral Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Feng Li
- Department of Oral Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Dian Xu
- State Key Laboratory of Oral and 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, China
| | - Jian Liu
- Department of Oral Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Jian Li
- Department of Oral Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Lingmei Zhong
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanshan Liu
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Dental Digital Medicine and 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
| | - Na Bai
- Department of Oral Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Wu S, Luo S, Cen Z, Li Q, Li L, Li W, Huang Z, He W, Liang G, Wu D, Zhou M, Li Y. All-in-one porous membrane enables full protection in guided bone regeneration. Nat Commun 2024; 15:119. [PMID: 38168072 PMCID: PMC10762214 DOI: 10.1038/s41467-023-43476-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 11/10/2023] [Indexed: 01/05/2024] Open
Abstract
The sophisticated hierarchical structure that precisely combines contradictory mechanical and biological characteristics is ideal for biomaterials, but it is challenging to achieve. Herein, we engineer a spatiotemporally hierarchical guided bone regeneration (GBR) membrane by rational bilayer integration of densely porous N-halamine functionalized bacterial cellulose nanonetwork facing the gingiva and loosely porous chitosan-hydroxyapatite composite micronetwork facing the alveolar bone. Our GBR membrane asymmetrically combine stiffness and flexibility, ingrowth barrier and ingrowth guiding, as well as anti-bacteria and cell-activation. The dense layer has a mechanically matched space maintenance capacity toward gingiva, continuously blocks fibroblasts, and prevents bacterial invasion with multiple mechanisms including release-killing, contact-killing, anti-adhesion, and nanopore-blocking; the loose layer is ultra-soft to conformally cover bone surfaces and defect cavity edges, enables ingrowth of osteogenesis-associated cells, and creates a favorable osteogenic microenvironment. As a result, our all-in-one porous membrane possesses full protective abilities in GBR.
Collapse
Affiliation(s)
- Shuyi Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 510055, Guangzhou, P. R. China
| | - Shulu Luo
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 510055, Guangzhou, P. R. China
| | - Zongheng Cen
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Chemistry, Sun Yat-sen University, 510006, Guangzhou, P.R. China
| | - Qianqian Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 510055, Guangzhou, P. R. China
| | - Luwei Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 510055, Guangzhou, P. R. China
| | - Weiran Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 510055, Guangzhou, P. R. China
| | - Zhike Huang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, P. R. China
| | - Wenyi He
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Chemistry, Sun Yat-sen University, 510006, Guangzhou, P.R. China
| | - Guobin Liang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 510055, Guangzhou, P. R. China
| | - Dingcai Wu
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Chemistry, Sun Yat-sen University, 510006, Guangzhou, P.R. China.
| | - Minghong Zhou
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, P. R. China.
| | - Yan Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 510055, Guangzhou, P. R. China.
| |
Collapse
|
28
|
Li R, Zhu Z, Zhang B, Jiang T, Zhu C, Mei P, Jin Y, Wang R, Li Y, Guo W, Liu C, Xia L, Fang B. Manganese Enhances the Osteogenic Effect of Silicon-Hydroxyapatite Nanowires by Targeting T Lymphocyte Polarization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305890. [PMID: 38039434 PMCID: PMC10811488 DOI: 10.1002/advs.202305890] [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: 08/20/2023] [Revised: 10/17/2023] [Indexed: 12/03/2023]
Abstract
Biomaterials encounter considerable challenges in extensive bone defect regeneration. The amelioration of outcomes may be attainable through the orchestrated modulation of both innate and adaptive immunity. Silicon-hydroxyapatite, for instance, which solely focuses on regulating innate immunity, is inadequate for long-term bone regeneration. Herein, extra manganese (Mn)-doping is utilized for enhancing the osteogenic ability by mediating adaptive immunity. Intriguingly, Mn-doping engenders heightened recruitment of CD4+ T cells to the bone defect site, concurrently manifesting escalated T helper (Th) 2 polarization and an abatement in Th1 cell polarization. This consequential immune milieu yields a collaborative elevation of interleukin 4, secreted by Th2 cells, coupled with attenuated interferon gamma, secreted by Th1 cells. This orchestrated interplay distinctly fosters the osteogenesis of bone marrow stromal cells and effectuates consequential regeneration of the mandibular bone defect. The modulatory mechanism of Th1/Th2 balance lies primarily in the indispensable role of manganese superoxide dismutase (MnSOD) and the phosphorylation of adenosine 5'-monophosphate-activated protein kinase (AMPK). In conclusion, this study highlights the transformative potential of Mn-doping in amplifying the osteogenic efficacy of silicon-hydroxyapatite nanowires by regulating T cell-mediated adaptive immunity via the MnSOD/AMPK pathway, thereby creating an anti-inflammatory milieu favorable for bone regeneration.
Collapse
Affiliation(s)
- Ruomei Li
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Zhiyu Zhu
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Bolin Zhang
- Department of StomatologyXinHua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai Jiao Tong University1665 Kongjiang RoadShanghai200092China
| | - Ting Jiang
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Cheng Zhu
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Peng Mei
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Yu Jin
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Ruiqing Wang
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Yixin Li
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Weiming Guo
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Chengxiao Liu
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Lunguo Xia
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| | - Bing Fang
- Department of OrthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University500 Quxi RoadShanghai200011China
| |
Collapse
|
29
|
El-Kady AM, Mahmoud EM, Sayed M, Kamel SM, Naga SM. In-vitro and in-vivo evaluation for the bio-natural Alginate/nano-Hydroxyapatite (Alg/n-HA) injectable hydrogel for critical size bone substitution. Int J Biol Macromol 2023; 253:126618. [PMID: 37659491 DOI: 10.1016/j.ijbiomac.2023.126618] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
Currently, bio-natural injectable hydrogels are receiving a lot of attention due to their ability to control, adjust, and adapt to random bone defects, in addition, to their ability to mimic the composition of natural bones. From such a viewpoint, this study goal is to prepare and characterize the injectable hydrogels paste based on the natural alginate (Alg) derived from brown sea algae as a polysaccharide polymer, which coupled with nano biogenic-hydroxyapatite (n-HA) prepared from eggshells and enriched with valuable trace elements. The viscosity and mechanical properties of the paste were investigated. As well as the in-vitro study in terms of water absorption and biodegradability in the PBS, biocompatibility and the capability of the injectable Alginate/n-Hydroxyapatite (Alg/n-HA) to regenerate bone for the most suitable injectable form. The injectable hydrogel (BP -B sample) was chosen for the study as it had an appropriate setting time for injecting (13 mins), and suitable compressive strength reached 6.3 MPa. The in vivo study was also carried out including a post-surgery follow-up test of the newly formed bone (NB) in the defect area after 10 and 20 weeks using different techniques such as (SEM/EDX) and histological analysis, the density of the newly formed bone by Dual x-ray absorptiometry (DEXA), blood biochemistry and the radiology test. The results proved that the injectable hydrogels Alginate/n-Hydroxyapatite (Alg/n-HA) had an appreciated biodegradability and bioactivity, which allow the progress of angiogenesis, endochondral ossification, and osteogenesis throughout the defect area, which positively impacts the healing time and ensures the full restoration for the well-mature bone tissue that similar to the natural bone.
Collapse
Affiliation(s)
- Abeer M El-Kady
- Glass Research Department, National Research Centre, El-Bohous Str., 12622 Cairo, Egypt
| | - E M Mahmoud
- Ceramics Department, National Research Centre, El-Bohous Str., 12622 Cairo, Egypt.
| | - M Sayed
- Ceramics Department, National Research Centre, El-Bohous Str., 12622 Cairo, Egypt
| | - S M Kamel
- Oral Biology Department, MSA University, Egypt
| | - S M Naga
- Ceramics Department, National Research Centre, El-Bohous Str., 12622 Cairo, Egypt
| |
Collapse
|
30
|
Bai L, Song P, Su J. Bioactive elements manipulate bone regeneration. BIOMATERIALS TRANSLATIONAL 2023; 4:248-269. [PMID: 38282709 PMCID: PMC10817798 DOI: 10.12336/biomatertransl.2023.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 01/30/2024]
Abstract
While bone tissue is known for its inherent regenerative abilities, various pathological conditions and trauma can disrupt its meticulously regulated processes of bone formation and resorption. Bone tissue engineering aims to replicate the extracellular matrix of bone tissue as well as the sophisticated biochemical mechanisms crucial for effective regeneration. Traditionally, the field has relied on external agents like growth factors and pharmaceuticals to modulate these processes. Although efficacious in certain scenarios, this strategy is compromised by limitations such as safety issues and the transient nature of the compound release and half-life. Conversely, bioactive elements such as zinc (Zn), magnesium (Mg) and silicon (Si), have garnered increasing interest for their therapeutic benefits, superior stability, and reduced biotic risks. Moreover, these elements are often incorporated into biomaterials that function as multifaceted bioactive components, facilitating bone regeneration via release on-demand. By elucidating the mechanistic roles and therapeutic efficacy of the bioactive elements, this review aims to establish bioactive elements as a robust and clinically viable strategy for advanced bone regeneration.
Collapse
Affiliation(s)
- Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
31
|
Ma Y, Wang S, Wang H, Chen X, Shuai Y, Wang H, Mao Y, He F. Mesenchymal stem cells and dental implant osseointegration during aging: from mechanisms to therapy. Stem Cell Res Ther 2023; 14:382. [PMID: 38124153 PMCID: PMC10734190 DOI: 10.1186/s13287-023-03611-1] [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: 08/29/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
Dental implants are widely used to replace missing teeth, providing patients with unparalleled levels of effectiveness, convenience, and affordability. The biological basis for the clinical success of dental implants is osseointegration. Bone aging is a high-risk factor for the reduced osseointegration and survival rates of dental implants. In aged individuals, mesenchymal stem cells (MSCs) in the bone marrow show imbalanced differentiation with a reduction in osteogenesis and an increase in adipogenesis. This leads to impaired osseointegration and implant failure. This review focuses on the molecular mechanisms underlying the dysfunctional differentiation of aged MSCs, which primarily include autophagy, transcription factors, extracellular vesicle secretion, signaling pathways, epigenetic modifications, microRNAs, and oxidative stress. Furthermore, this review addresses the pathological changes in MSCs that affect osseointegration and discusses potential therapeutic interventions to enhance osseointegration by manipulating the mechanisms underlying MSC aging.
Collapse
Affiliation(s)
- Yang Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Siyuan Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Hui Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiaoyu Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yi Shuai
- Nanjing Jinling Hospital: East Region Military Command General Hospital, Nanjing, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| | - Yingjie Mao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| | - Fuming He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| |
Collapse
|
32
|
Wu Y, Huo S, Liu S, Hong Q, Wang Y, Lyu Z. Cu-Sr Bilayer Bioactive Glass Nanoparticles/Polydopamine Functionalized Polyetheretherketone Enhances Osteogenic Activity and Prevents Implant-Associated Infections through Spatiotemporal Immunomodulation. Adv Healthc Mater 2023; 12:e2301772. [PMID: 37723927 DOI: 10.1002/adhm.202301772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/03/2023] [Indexed: 09/20/2023]
Abstract
Key factors contributing to implantation failures include implant-associated infections (IAIs) and insufficient osseointegration of the implants. Polyetheretherketone (PEEK) is widely used in orthopedics, yet its clinical applications are restricted due to its poor osteogenic and antibacterial properties as well as inadequate immune responses. To overcome these drawbacks, a novel spatiotemporal immunomodulation approach is proposed, chelating Cu-Sr bilayer bioactive glass nanoparticles (CS-BGNs) onto the PEEK surface via polydopamine (PDA). The CS-BGNs possess a bilayer core-shell structure where copper is distributed in the outer layer and strontium is clustered in the inner layer. The results show that CS-BGNs/PDA functionalized PEEK demonstrates a controlled and sequential release of Cu2+ and Sr2+ . In the early stage, Cu2+ from the outer layer releases rapidly, while Sr2+ from the inner layer releases in the late stage. This well-ordered release pattern modulates the phenotypic transition of macrophages, which induces M1 polarization in the early stage and M2 polarization in the late stage. Combined with the direct effects of Cu2+ and Sr2+ , the spatiotemporal immunomodulation not only benefits the early antibacterial and tissue-healing process, but also promotes the long-term process of osseointegration, providing new perspectives on the design of novel immunomodulatory biomaterials.
Collapse
Affiliation(s)
- Yuezhou Wu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Shicheng Huo
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Shu Liu
- Department of Orthopedic Surgery, Changhai Hospital, The Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Qimin Hong
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China
| | - You Wang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Zhuocheng Lyu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China
| |
Collapse
|
33
|
Liu X, Zhou Z, Zeng WN, Zeng Q, Zhang X. The role of toll-like receptors in orchestrating osteogenic differentiation of mesenchymal stromal cells and osteoimmunology. Front Cell Dev Biol 2023; 11:1277686. [PMID: 37941898 PMCID: PMC10629627 DOI: 10.3389/fcell.2023.1277686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Osteoimmunology is a concept involving molecular and cellular crosstalk between the skeletal and immune systems. Toll-like receptors (TLRs) are widely expressed both on mesenchymal stromal cells (MSCs), the hematopoietic cells, and immune cells in the osteogenic microenvironment for bone development or repair. TLRs can sense both exogenous pathogen-associated molecular patterns (PAMPs) derived from microorganisms, and damage-associated molecular patterns (DAMPs) derived from normal cells subjected to injury, inflammation, or cell apoptosis under physiological or pathological conditions. Emerging studies reported that TLR signaling plays an important role in bone remodeling by directly impacting MSC osteogenic differentiation or osteoimmunology. However, how to regulate TLR signaling is critical and remains to be elucidated to promote the osteogenic differentiation of MSCs and new bone formation for bone tissue repair. This review outlines distinct TLR variants on MSCs from various tissues, detailing the impact of TLR pathway activation or inhibition on MSC osteogenic differentiation. It also elucidates TLR pathways' interplay with osteoclasts, immune cells, and extracellular vesicles (EVs) derived from MSCs. Furthermore, we explore biomaterial-based activation to guide MSCs' osteogenic differentiation. Therefore, understanding TLRs' role in this context has significant implications for advancing bone regeneration and repair strategies.
Collapse
Affiliation(s)
- Xiaoyang Liu
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Zongke Zhou
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Wei-Nan Zeng
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Zeng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterials & Institute of Regulatory Science for Medical Devices & NMPA Research Base of Regulatory Science for Medical Devices, Sichuan University, Chengdu, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterials & Institute of Regulatory Science for Medical Devices & NMPA Research Base of Regulatory Science for Medical Devices, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Hao S, Wang M, Yin Z, Jing Y, Bai L, Su J. Microenvironment-targeted strategy steers advanced bone regeneration. Mater Today Bio 2023; 22:100741. [PMID: 37576867 PMCID: PMC10413201 DOI: 10.1016/j.mtbio.2023.100741] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Treatment of large bone defects represents a great challenge in orthopedic and craniomaxillofacial surgery. Traditional strategies in bone tissue engineering have focused primarily on mimicking the extracellular matrix (ECM) of bone in terms of structure and composition. However, the synergistic effects of other cues from the microenvironment during bone regeneration are often neglected. The bone microenvironment is a sophisticated system that includes physiological (e.g., neighboring cells such as macrophages), chemical (e.g., oxygen, pH), and physical factors (e.g., mechanics, acoustics) that dynamically interact with each other. Microenvironment-targeted strategies are increasingly recognized as crucial for successful bone regeneration and offer promising solutions for advancing bone tissue engineering. This review provides a comprehensive overview of current microenvironment-targeted strategies and challenges for bone regeneration and further outlines prospective directions of the approaches in construction of bone organoids.
Collapse
Affiliation(s)
- Shuyue Hao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, China
| |
Collapse
|
35
|
Sivakumar PM, Yetisgin AA, Demir E, Sahin SB, Cetinel S. Polysaccharide-bioceramic composites for bone tissue engineering: A review. Int J Biol Macromol 2023; 250:126237. [PMID: 37567538 DOI: 10.1016/j.ijbiomac.2023.126237] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/05/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023]
Abstract
Limitations associated with conventional bone substitutes such as autografts, increasing demand for bone grafts, and growing elderly population worldwide necessitate development of unique materials as bone graft substitutes. Bone tissue engineering (BTE) would ensure therapy advancement, efficiency, and cost-effective treatment modalities of bone defects. One way of engineering bone tissue scaffolds by mimicking natural bone tissue composed of organic and inorganic phases is to utilize polysaccharide-bioceramic hybrid composites. Polysaccharides are abundant in nature, and present in human body. Biominerals, like hydroxyapatite are present in natural bone and some of them possess osteoconductive and osteoinductive properties. Ion doped bioceramics could substitute protein-based biosignal molecules to achieve osteogenesis, vasculogenesis, angiogenesis, and stress shielding. This review is a systemic summary on properties, advantages, and limitations of polysaccharide-bioceramic/ion doped bioceramic composites along with their recent advancements in BTE.
Collapse
Affiliation(s)
- Ponnurengam Malliappan Sivakumar
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Institute of Research and Development, Duy Tan University, Da Nang 550000, Viet Nam; School of Medicine and Pharmacy, Duy Tan University, Da Nang 550000, Viet Nam.
| | - Abuzer Alp Yetisgin
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Sabanci University, Faculty of Engineering and Natural Sciences, Materials Science and Nano-Engineering Program, Istanbul 34956, Turkey
| | - Ebru Demir
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Sabanci University, Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Istanbul 34956, Turkey
| | - Sevilay Burcu Sahin
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Sabanci University, Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Istanbul 34956, Turkey
| | - Sibel Cetinel
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Sabanci University, Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Istanbul 34956, Turkey.
| |
Collapse
|
36
|
Gu G, Li Y, Zuo K, Xiao G. The Influence of pH Value on the Microstructure and Properties of Strontium Phosphate Chemical Conversion Coatings on Titanium. Molecules 2023; 28:6651. [PMID: 37764427 PMCID: PMC10535797 DOI: 10.3390/molecules28186651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Strontium (Sr) is a trace element in the human body that can promote bone formation and inhibit bone absorption. A conversion coating of strontium phosphate (Sr-P) on the surface of titanium (Ti) can improve its biological properties and has many potential applications in the fields of dentistry and orthopedics. In the present study, Sr-P coatings with SrHPO4 and Sr3(PO4)2 crystals on Ti are prepared by a phosphate chemical conversion (PCC) treatment and the effect of pH values on the properties of the Sr-P coatings is researched. The results prove that the phase composition, morphology, and corrosion resistance of the coated Ti vary according to the pH values of the PCC solution. The morphology of the conversion deposition on Ti changes from plat-like to cluster-like and then to homogeneous microcrystals as the pH value changes from 2.50 to 3.25. Only discrete SrHPO4 crystals are generated on the substrate at lower pH values, while relatively stable Sr3(PO4)2 and SrHPO4 crystals grow and subsequently form an integrated coating on the Ti as the pH exceeds 2.50. The cross-sectional morphologies and bonding strength of different coatings are also researched. The corrosion resistance of coated Ti improves compared with that of bare Ti because of the Sr-P coatings with a Sr3(PO4)2 phase. In addition, it is indicated that the Sr-P coatings on Ti can improve the adhesion and differentiation of BMSCs.
Collapse
Affiliation(s)
- Guochao Gu
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250061, China; (G.G.); (Y.L.); (K.Z.)
- School of Materials Science and Engineering, Shandong University, Jinan 250061, China
| | - Yibo Li
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250061, China; (G.G.); (Y.L.); (K.Z.)
- School of Materials Science and Engineering, Shandong University, Jinan 250061, China
| | - Kangqing Zuo
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250061, China; (G.G.); (Y.L.); (K.Z.)
- School of Materials Science and Engineering, Shandong University, Jinan 250061, China
| | - Guiyong Xiao
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250061, China; (G.G.); (Y.L.); (K.Z.)
- School of Materials Science and Engineering, Shandong University, Jinan 250061, China
| |
Collapse
|
37
|
Zhang Y, Zhang W, Zhang X, Zhou Y. Erbium-ytterbium containing upconversion mesoporous bioactive glass microspheres for tissue engineering: luminescence monitoring of biomineralization and drug release. Acta Biomater 2023; 168:628-636. [PMID: 37454706 DOI: 10.1016/j.actbio.2023.07.014] [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: 04/24/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
The development of functional biomaterials with real-time monitoring of mineralization processes, drug release and biodistribution has potential applications but remains an unsolved challenge. Herein, erbium- and ytterbium- containing mesoporous bioactive glass microspheres (MBGs:Er/Yb) with blue and red emission at an excitation wavelength of 980 nm were synthesized by a sol-gel combined with organic template method. As the concentration of Yb3+ ions gradually increases, the emission intensity of the MBGs:Er/Yb exhibits a clear concentration quenching effect. Combined with in vitro bioactivity tests, the optimal molar ratio of Er3+/Yb3+ was determined to be 4:3. Therefore, MBGs:4Er/3Yb was selected for in vitro biomineralization and drug release monitoring. The results of biomineralization monitoring show that the upconversion luminescence intensity is closely related to the degree of biomineralization. The upconversion luminescence intensity of MBGs:4Er/3Yb is quenched with the increase of the degree of biomineralization. The degree of luminescence quenching during biomineralization can be semiquantized. Drug release monitoring experiments showed that the anticancer drug doxorubicin hydrochloride (DOX) was successfully loaded into MBGs:4Er/3Yb and selectively quenched the green emission. When DOX was released, the green emission recovered stably, and It/I0 increased gradually. Moreover, there was a linear relationship between It/I0 and cumulative drug release, indicating that DOX-MBGs:4Er/3Yb is highly sensitive to DOX release, and monitoring the It/I0 values of DOX-MBGs:4Er/3Yb can achieve real-time tracking of the DOX release process to a certain extent. In conclusion, MBGs:4Er/3Yb has potential application as an upconversion luminescence biomonitoring material in the field of bone tissue engineering. STATEMENT OF SIGNIFICANCE: Mesoporous bioactive glasses have great potential for applications in bone tissue repair due to their excellent biological properties, but the effective information of the repair process cannot be grasped in a timely manner. Therefore, real-time monitoring of mineralization and drug release processes will be beneficial to obtain the degree of healing and optimize the amount and distribution of drugs to improve targeted therapeutic effects. For biomaterials, in vitro biological properties determine their biological properties in vivo, where the environment is more complex and diverse, and thus in vitro biomonitoring is particularly crucial. The organic combination of physical properties and biological properties will also provide a feasible idea for the development of biomaterials.
Collapse
Affiliation(s)
- Ying Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China.
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Xiaona Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Yu Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| |
Collapse
|
38
|
Yang Z, Wang B, Liu W, Li X, Liang K, Fan Z, Li JJ, Niu Y, He Z, Li H, Wang D, Lin J, Du Y, Lin J, Xing D. In situ self-assembled organoid for osteochondral tissue regeneration with dual functional units. Bioact Mater 2023; 27:200-215. [PMID: 37096194 PMCID: PMC10121637 DOI: 10.1016/j.bioactmat.2023.04.002] [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: 02/12/2023] [Revised: 04/01/2023] [Accepted: 04/02/2023] [Indexed: 04/26/2023] Open
Abstract
The regeneration of hierarchical osteochondral units is challenging due to difficulties in inducing spatial, directional and controllable differentiation of mesenchymal stem cells (MSCs) into cartilage and bone compartments. Emerging organoid technology offers new opportunities for osteochondral regeneration. In this study, we developed gelatin-based microcryogels customized using hyaluronic acid (HA) and hydroxyapatite (HYP), respectively for inducing cartilage and bone regeneration (denoted as CH-Microcryogels and OS-Microcryogels) through in vivo self-assembly into osteochondral organoids. The customized microcryogels showed good cytocompatibility and induced chondrogenic and osteogenic differentiation of MSCs, while also demonstrating the ability to self-assemble into osteochondral organoids with no delamination in the biphasic cartilage-bone structure. Analysis by mRNA-seq showed that CH-Microcryogels promoted chondrogenic differentiation and inhibited inflammation, while OS-Microcryogels facilitated osteogenic differentiation and suppressed the immune response, by regulating specific signaling pathways. Finally, the in vivo engraftment of pre-differentiated customized microcryogels into canine osteochondral defects resulted in the spontaneous assembly of an osteochondral unit, inducing simultaneous regeneration of both articular cartilage and subchondral bone. In conclusion, this novel approach for generating self-assembling osteochondral organoids utilizing tailor-made microcryogels presents a highly promising avenue for advancing the field of tissue engineering.
Collapse
Affiliation(s)
- Zhen Yang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Bin Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 10081, China
| | - Xiaoke Li
- Department of Orthopedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Zejun Fan
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, Australia
| | - Yudi Niu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Zihao He
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Hui Li
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Jianhao Lin
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Dan Xing
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| |
Collapse
|
39
|
Dutta SD, Ganguly K, Hexiu J, Randhawa A, Moniruzzaman M, Lim KT. A 3D Bioprinted Nanoengineered Hydrogel with Photoactivated Drug Delivery for Tumor Apoptosis and Simultaneous Bone Regeneration via Macrophage Immunomodulation. Macromol Biosci 2023; 23:e2300096. [PMID: 37087681 DOI: 10.1002/mabi.202300096] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/12/2023] [Indexed: 04/24/2023]
Abstract
One of the significant challenges in bone tissue engineering (BTE) is the healing of traumatic tissue defects owing to the recruitment of local infection and delayed angiogenesis. Herein, a 3D printable multi-functional hydrogel composing polyphenolic carbon quantum dots (CQDs, 100 µg mL-1 ) and gelatin methacryloyl (GelMA, 12 wt%) is reported for robust angiogenesis, bone regeneration and anti-tumor therapy. The CQDs are synthesized from a plant-inspired bioactive molecule, 1, 3, 5-trihydroxybenzene. The 3D printed GelMA-CQDs hydrogels display typical shear-thinning behavior with excellent printability. The fabricated hydrogel displayed M2 polarization of macrophage (Raw 264.7) cells via enhancing anti-inflammatory genes (e.g., IL-4 and IL10), and induced angiogenesis and osteogenesis of human bone mesenchymal stem cells (hBMSCs). The bioprinted hBMSCs are able to produce vessel-like structures after 14 d of incubation. Furthermore, the 3D printed hydrogel scaffolds also show remarkable near infra-red (NIR) responsive properties under 808 nm NIR light (1.0 W cm-2 ) irradiation with controlled release of antitumor drugs (≈49%) at pH 6.5, and thereby killing the osteosarcoma cells. Therefore, it is anticipated that the tissue regeneration and healing ability with therapeutic potential of the GelMA-CQDs scaffolds may provide a promising alternative for traumatic tissue regeneration via augmenting angiogenesis and accelerated immunomodulation.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jin Hexiu
- Department of Oral and Maxillofacial Surgery, Capital Medical University, Beijing, China
| | - Aayushi Randhawa
- Department of Biosystems Engineering, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 100069, Republic of Korea
| | - Md Moniruzzaman
- Department of Chemical and Biological Engineering, Gachon University, Seongnam, 1342, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 100069, Republic of Korea
| |
Collapse
|
40
|
Wu P, Shen L, Liu HF, Zou XH, Zhao J, Huang Y, Zhu YF, Li ZY, Xu C, Luo LH, Luo ZQ, Wu MH, Cai L, Li XK, Wang ZG. The marriage of immunomodulatory, angiogenic, and osteogenic capabilities in a piezoelectric hydrogel tissue engineering scaffold for military medicine. Mil Med Res 2023; 10:35. [PMID: 37525300 PMCID: PMC10388535 DOI: 10.1186/s40779-023-00469-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/05/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Most bone-related injuries to grassroots troops are caused by training or accidental injuries. To establish preventive measures to reduce all kinds of trauma and improve the combat effectiveness of grassroots troops, it is imperative to develop new strategies and scaffolds to promote bone regeneration. METHODS In this study, a porous piezoelectric hydrogel bone scaffold was fabricated by incorporating polydopamine (PDA)-modified ceramic hydroxyapatite (PDA-hydroxyapatite, PHA) and PDA-modified barium titanate (PDA-BaTiO3, PBT) nanoparticles into a chitosan/gelatin (Cs/Gel) matrix. The physical and chemical properties of the Cs/Gel/PHA scaffold with 0-10 wt% PBT were analyzed. Cell and animal experiments were performed to characterize the immunomodulatory, angiogenic, and osteogenic capabilities of the piezoelectric hydrogel scaffold in vitro and in vivo. RESULTS The incorporation of BaTiO3 into the scaffold improved its mechanical properties and increased self-generated electricity. Due to their endogenous piezoelectric stimulation and bioactive constituents, the as-prepared Cs/Gel/PHA/PBT hydrogels exhibited cytocompatibility as well as immunomodulatory, angiogenic, and osteogenic capabilities; they not only effectively induced macrophage polarization to M2 phenotype but also promoted the migration, tube formation, and angiogenic differentiation of human umbilical vein endothelial cells (HUVECs) and facilitated the migration, osteo-differentiation, and extracellular matrix (ECM) mineralization of MC3T3-E1 cells. The in vivo evaluations showed that these piezoelectric hydrogels with versatile capabilities significantly facilitated new bone formation in a rat large-sized cranial injury model. The underlying molecular mechanism can be partly attributed to the immunomodulation of the Cs/Gel/PHA/PBT hydrogels as shown via transcriptome sequencing analysis, and the PI3K/Akt signaling axis plays an important role in regulating macrophage M2 polarization. CONCLUSION The piezoelectric Cs/Gel/PHA/PBT hydrogels developed here with favorable immunomodulation, angiogenesis, and osteogenesis functions may be used as a substitute in periosteum injuries, thereby offering the novel strategy of applying piezoelectric stimulation in bone tissue engineering for the enhancement of combat effectiveness in grassroots troops.
Collapse
Affiliation(s)
- Ping Wu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Lin Shen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
| | - Hui-Fan Liu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiang-Hui Zou
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Juan Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
| | - Yu Huang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
| | - Yu-Fan Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhao-Yu Li
- Department of Overseas Education College, Jimei University, Xiamen, 361021, Fujian, China
| | - Chao Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Li-Hua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zhi-Qiang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Min-Hao Wu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Xiao-Kun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Zhou-Guang Wang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
41
|
Pan X, Ou M, Lu Y, Nie Q, Dai X, Liu O. Immunomodulatory zinc-based materials for tissue regeneration. BIOMATERIALS ADVANCES 2023; 152:213503. [PMID: 37331243 DOI: 10.1016/j.bioadv.2023.213503] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/20/2023]
Abstract
Zinc(Zn)-based materials have contributed greatly to the rapid advancements in tissue engineering. The qualities they possess that make them so beneficial include their excellent biodegradability, biocompatibility, anti-bacterial activity, among and several others. Biomedical materials that act as a foreign body, will inevitably cause host immune response when introduced to the human body. As the osteoimmunology develops, the immunomodulatory characteristics of biomaterials have become an appealing concept to improve implant-tissue interaction and tissue restoration. Recently, Zn-based materials have also displayed immunomodulatory functions, especially macrophage polarization states. It can promote the transformation of M1 macrophages into M2 macrophages to enhance the tissue regeneration and reconstruction. This review covers mainly Zn-based materials and their characteristics, including metallic Zn alloys and Zn ceramics. We highlight the current advancements in the type of immune responses, as well as the mechanisms, that are induced by Zn-based biomaterials, most importantly the regulation of innate immunity and the mechanism of promoting tissue regeneration. To this end, we discuss their applications in biomedicine, and conclude with an outlook on future research challenges.
Collapse
Affiliation(s)
- Xiaoman Pan
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410005, China
| | - Mingning Ou
- Xiangya Hospital & Xiangya School of Medicine, Central South University, Changsha 410005, China
| | - Yixuan Lu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410005, China
| | - Qian Nie
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410005, China
| | - Xiaohan Dai
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410005, China.
| | - Ousheng Liu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410005, China.
| |
Collapse
|
42
|
Fosca M, Streza A, Antoniac IV, Vadalà G, Rau JV. Ion-Doped Calcium Phosphate-Based Coatings with Antibacterial Properties. J Funct Biomater 2023; 14:jfb14050250. [PMID: 37233360 DOI: 10.3390/jfb14050250] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Ion-substituted calcium phosphate (CP) coatings have been extensively studied as promising materials for biomedical implants due to their ability to enhance biocompatibility, osteoconductivity, and bone formation. This systematic review aims to provide a comprehensive analysis of the current state of the art in ion-doped CP-based coatings for orthopaedic and dental implant applications. Specifically, this review evaluates the effects of ion addition on the physicochemical, mechanical, and biological properties of CP coatings. The review also identifies the contribution and additional effects (in a separate or a synergistic way) of different components used together with ion-doped CP for advanced composite coatings. In the final part, the effects of antibacterial coatings on specific bacteria strains are reported. The present review could be of interest to researchers, clinicians, and industry professionals involved in the development and application of CP coatings for orthopaedic and dental implants.
Collapse
Affiliation(s)
- Marco Fosca
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere 100, 00133 Rome, Italy
| | - Alexandru Streza
- Faculty of Material Science and Engineering, University Politehnica of Bucharest, 313 Splaiul Independentei Street, District 6, 060042 Bucharest, Romania
| | - Iulian V Antoniac
- Faculty of Material Science and Engineering, University Politehnica of Bucharest, 313 Splaiul Independentei Street, District 6, 060042 Bucharest, Romania
- Academy of Romanian Scientists, 54 Splaiul Independentei Street, District 5, 050094 Bucharest, Romania
| | - Gianluca Vadalà
- Laboratory of Regenerative Orthopaedics, Research Unit of Orthopaedic, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy
- Operative Research Unit of Orthopaedics, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Julietta V Rau
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere 100, 00133 Rome, Italy
| |
Collapse
|
43
|
Wen Z, Shi X, Li X, Liu W, Liu Y, Zhang R, Yu Y, Su J. Mesoporous TiO 2 Coatings Regulate ZnO Nanoparticle Loading and Zn 2+ Release on Titanium Dental Implants for Sustained Osteogenic and Antibacterial Activity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:15235-15249. [PMID: 36926829 DOI: 10.1021/acsami.3c00812] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Two major issues are currently hindering the clinical practice of titanium dental implants for the lack of biological activities: immediate/early loading risks and peri-implantitis. To solve these issues, it is urgent to develop multifunctional implants modified with effective osteogenic and antibacterial properties. Zinc oxide nanoparticles (ZnO NPs) possess superior antibacterial activity; however, they can rapidly release Zn2+, causing cytotoxicity. In this study, a potential dental implant modification was creatively developed as ZnO nanoparticle-loaded mesoporous TiO2 coatings (nZnO/MTC-Ti) via the evaporation-induced self-assembly method (EISA) and one-step spin coating. The mesoporous TiO2 coatings (MTCs) regulated the synthesis and loading of ZnO NPs inside the nanosized pores. The synergistic effects of MTC and ZnO NPs on nZnO/MTC-Ti not only controlled the long-term steady-state release of Zn2+ but also optimized the charge distribution on the surface. Therefore, the cytotoxicity of ZnO NPs was resolved without triggering excessive reactive oxygen species (ROS). The increased extracellular Zn2+ further promoted a favorable intracellular zinc ion microenvironment through the modulation of zinc transporters (ZIP1 and ZnT1). Owing to that, the adhesion, proliferation, and osteogenic activity of bone mesenchymal stem cells (BMSCs) were improved. Additionally, nZnO/MTC-Ti inhibited the proliferation of oral pathogens (Pg and Aa) by inducing bacterial ROS production. For in vivo experiments, different implants were implanted into the alveolar fossa of Sprague-Dawley rats immediately after tooth extraction. The nZnO/MTC-Ti implants were found to possess a higher capability for enhancing bone regeneration, antibiosis, and osseointegration in vivo. These findings suggested the outstanding performance of nZnO/MTC-Ti implants in accelerating osseointegration and inhibiting bacterial infection, indicating a huge potential for solving immediate/early loading risks and peri-implantitis of dental implants.
Collapse
Affiliation(s)
- Zhuo Wen
- Department of Prosthodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, P. R. China
| | - Xinyue Shi
- Institute of New Energy for Vehicles, Shanghai Key Laboratory for Development and Application of Metallic Functional Materials, School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Xuejing Li
- Department of Prosthodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, P. R. China
| | - Weicai Liu
- Department of Prosthodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, P. R. China
| | - Yukun Liu
- Institute of New Energy for Vehicles, Shanghai Key Laboratory for Development and Application of Metallic Functional Materials, School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Renyuan Zhang
- Institute of New Energy for Vehicles, Shanghai Key Laboratory for Development and Application of Metallic Functional Materials, School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Yiqiang Yu
- Department of Prosthodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, P. R. China
| | - Jiansheng Su
- Department of Prosthodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, P. R. China
| |
Collapse
|
44
|
Liu X, Huang H, Zhang J, Sun T, Zhang W, Li Z. Recent Advance of Strontium Functionalized in Biomaterials for Bone Regeneration. Bioengineering (Basel) 2023; 10:bioengineering10040414. [PMID: 37106601 PMCID: PMC10136039 DOI: 10.3390/bioengineering10040414] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Bone defect disease causes damage to people’s lives and property, and how to effectively promote bone regeneration is still a big clinical challenge. Most of the current repair methods focus on filling the defects, which has a poor effect on bone regeneration. Therefore, how to effectively promote bone regeneration while repairing the defects at the same time has become a challenge for clinicians and researchers. Strontium (Sr) is a trace element required by the human body, which mainly exists in human bones. Due to its unique dual properties of promoting the proliferation and differentiation of osteoblasts and inhibiting osteoclast activity, it has attracted extensive research on bone defect repair in recent years. With the deep development of research, the mechanisms of Sr in the process of bone regeneration in the human body have been clarified, and the effects of Sr on osteoblasts, osteoclasts, mesenchymal stem cells (MSCs), and the inflammatory microenvironment in the process of bone regeneration have been widely recognized. Based on the development of technology such as bioengineering, it is possible that Sr can be better loaded onto biomaterials. Even though the clinical application of Sr is currently limited and relevant clinical research still needs to be developed, Sr-composited bone tissue engineering biomaterials have achieved satisfactory results in vitro and in vivo studies. The Sr compound together with biomaterials to promote bone regeneration will be a development direction in the future. This review will present a brief overview of the relevant mechanisms of Sr in the process of bone regeneration and the related latest studies of Sr combined with biomaterials. The aim of this paper is to highlight the potential prospects of Sr functionalized in biomaterials.
Collapse
|
45
|
Paramasivan M, Sampath Kumar TS, Kanniyappan H, Muthuvijayan V, Chandra TS. Biomimetic ion substituted and Co-substituted hydroxyapatite nanoparticle synthesis using Serratia Marcescens. Sci Rep 2023; 13:4513. [PMID: 36934131 PMCID: PMC10024725 DOI: 10.1038/s41598-023-30996-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/06/2023] [Indexed: 03/20/2023] Open
Abstract
Biomimicry is becoming deep-rooted as part of bioceramics owing to its numerous functional advantages. Naturally occurring hydroxyapatite (HA) apart from primary nano structures are also characterised by various ionic substitutions. The ease of accommodating such key elements into the HA lattice is known to enhance bone healing properties of bioceramics. In this work, hydroxyapatite synthesized via biomimetic approach was substituted with individual as well as multiple cations for potential applications in bone repair. Ion substitutions of Sr, Mg and Zn was carried out on HA for the first time by using Serratia grown in a defined biomineralization medium. The individual ions of varying concentration substituted in Serratia HA (SHA) (Sr SHA, Mg SHA and Zn SHA) were analysed for crystallinity, functional groups, morphology and crystal size. All three showed decreased crystallinity, phase purity, large agglomerated aggregates and needle-shaped morphologies. Fourier transform infrared spectroscopy (FTIR) spectra indicated increased carbonate content of 5.8% resembling that of natural bone. Additionally, the reduced O-H intensities clearly portrayed disruption of HA lattice and subsequent ion-substitution. The novelty of this study lies primarily in investigating the co-substitution of a combination of 1% Sr, Zn and Mg in SHA and establishing the associated change in bone parameters. Scanning electron microscope (SEM) and transmission electron microscope (TEM) images clearly illustrated uniform nano-sized agglomerates of average dimensions of 20-50 nm length and 8-15 nm width for Sr SHA; 10-40 nm length and 8-10 nm width for both Zn SHA and Mg SHA and 40-70 nm length and 4-10 nm width in the case of 1% Sr, Zn, Mg SHA. In both individual as well as co-substitutions, significant peak shifts were not observed possibly due to the lower concentrations. However, cell volumes increased in both cases due to presence of Sr2+ validating its dominant integration into the SHA lattice. Rich trace ion deposition was presented by energy dispersive X-ray spectroscopy (EDS) and quantified using inductively coupled plasma optical emission spectrometer (ICP-OES). In vitro cytotoxicity studies in three cell lines viz. NIH/3T3 fibroblast cells, MG-63 osteosarcoma cells and RAW 264.7 macrophages showed more than 90% cell viability proving the biocompatible nature of 1% Sr, Zn and Mg in SHA. Microbial biomineralization by Serratia produced nanocrystals of HA that mimicked "bone-like apatite" as evidenced by pure phase, carbonated groups, reduced crystallinity, nano agglomerates, variations in cell parameters, rich ion deposition and non-toxic nature. Therefore ion-substituted and co-substituted biomineralized nano SHA appears to be a suitable candidate for applications in biomedicine addressing bone injuries and aiding regeneration as a result of its characteristics close to that of the human bone.
Collapse
Affiliation(s)
- Mareeswari Paramasivan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India
- Medical Materials Laboratory, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India
| | - T S Sampath Kumar
- Medical Materials Laboratory, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India.
| | - Hemalatha Kanniyappan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India
| | - Vignesh Muthuvijayan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India
| | - T S Chandra
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India.
| |
Collapse
|
46
|
Abstract
Collagen is commonly used as a regenerative biomaterial due to its excellent biocompatibility and wide distribution in tissues. Different kinds of hybridization or cross-links are favored to offer improvements to satisfy various needs of biomedical applications. Previous reviews have been made to introduce the sources and structures of collagen. In addition, biological and mechanical properties of collagen-based biomaterials, their modification and application forms, and their interactions with host tissues are pinpointed. However, there is still no review about collagen-based biomaterials for tissue engineering. Therefore, we aim to summarize and discuss the progress of collagen-based materials for tissue regeneration applications in this review. We focus on the utilization of collagen-based biomaterials for bones, cartilages, skin, dental, neuron, cornea, and urological applications and hope these experiences and outcomes can provide inspiration and practical techniques for the future development of collagen-based biomaterials in related application fields. Moreover, future improving directions and challenges for collagen-based biomaterials are proposed as well.
Collapse
Affiliation(s)
- Yiyu Wang
- Department of Prosthodontics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Zhengke Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Yan Dong
- Department of Prosthodontics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
47
|
Zhang Q, Xin M, Yang S, Wu Q, Xiang X, Wang T, Zhong W, Helder MN, Jaspers RT, Pathak JL, Xiao Y. Silica nanocarrier-mediated intracellular delivery of rapamycin promotes autophagy-mediated M2 macrophage polarization to regulate bone regeneration. Mater Today Bio 2023; 20:100623. [PMID: 37077506 PMCID: PMC10106556 DOI: 10.1016/j.mtbio.2023.100623] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/16/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
Targeting macrophages to regulate the immune microenvironment is a new strategy for bone regeneration with nano-drugs. Nano-drugs have achieved surprising anti-inflammatory and bone-regenerative effects, however, their underlying mechanisms in macrophages remain to be clarified. Macrophage polarization, immunomodulation, and osteogenesis are governed by autophagy. Rapamycin, an autophagy inducer, has shown promising results in bone regeneration, but high dose-mediated cytotoxicity and low bioavailability hinder its clinical application. This study aimed to develop rapamycin-loaded virus-like hollow silica nanoparticles (R@HSNs) which are easily phagocytosed by macrophages and translocated to lysosomes. R@HSNs induced macrophage autophagy, promoted M2 polarization, and alleviated the degree of M1 polarization as indicated by the downregulation of inflammatory factors IL-6, IL-1β, TNF-α, and iNOS, and upregulation of anti-inflammatory factors CD163, CD206, IL-1ra, IL-10, and TGF-β. These effects were nullified by cytochalasin B-induced inhibition of R@HSNs uptake in macrophages. The conditioned medium (CM) collected from R@HSNs-treated macrophages promoted osteogenic differentiation of mouse bone marrow mesenchymal stromal cells (mBMSCs). In a mouse calvaria defect model, free rapamycin treatment was inhibited, but R@HSNs robustly promoted bone defect healing. In conclusion, silica nanocarrier-mediated intracellular rapamycin delivery to macrophages effectively triggers autophagy-mediated M2 macrophage polarization, further enhancing bone regeneration by triggering osteogenic differentiation of mBMSCs.
Collapse
Affiliation(s)
- Qing Zhang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 BT Amsterdam, the Netherlands
| | - Mengyu Xin
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Shuang Yang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Qiuyu Wu
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Xi Xiang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Tianqi Wang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wen Zhong
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Marco N. Helder
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 HV Amsterdam, the Netherlands
| | - Richard T. Jaspers
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 BT Amsterdam, the Netherlands
| | - Janak Lal Pathak
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- Corresponding author.
| | - Yin Xiao
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- School of Medicine and Dentistry & Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, QLD 4000, Australia
- Corresponding author. Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China.
| |
Collapse
|
48
|
Three-Dimensional Printing of Poly-L-Lactic Acid Composite Scaffolds with Enhanced Bioactivity and Controllable Zn Ion Release Capability by Coupling with Carbon-ZnO. Bioengineering (Basel) 2023; 10:bioengineering10030307. [PMID: 36978698 PMCID: PMC10045836 DOI: 10.3390/bioengineering10030307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Poly-L-lactic acid (PLLA) has gained great popularity with researchers in regenerative medicine owing to its superior biocompatibility and biodegradability, although its inadequate bioactivity inhibits the further use of PLLA in the field of bone regeneration. Zinc oxide (ZnO) has been utilized to improve the biological performance of biopolymers because of its renowned osteogenic activity. However, ZnO nanoparticles tend to agglomerate in the polymer matrix due to high surface energy, which would lead to the burst release of the Zn ion and, thus, cytotoxicity. In this study, to address this problem, carbon–ZnO (C–ZnO) was first synthesized through the carbonization of ZIF-8. Then, C–ZnO was introduced to PLLA powder before it was manufactured as scaffolds (PLLA/C–ZnO) by a selective laser sintering 3D printing technique. The results showed that the PLLA/C–ZnO scaffold was able to continuously release Zn ions in a reasonable range, which can be attributed to the interaction of Zn–N bonding and the shielding action of the PLLA scaffold. The controlled release of Zn ions from the scaffold further facilitated cell adhesion and proliferation and improved the osteogenic differentiation ability at the same time. In addition, C–ZnO endowed the scaffold with favorable photodynamic antibacterial ability, which was manifested by an efficient antibacterial rate of over 95%.
Collapse
|
49
|
Wu J, Chi YQ, Yan YJ, Ji MZ, Chen X, Yang XQ, Gao Y, Zou Q, Zou L, Li XY. Investigation of the antibacterial effect, osteogenic activity, and tracing properties of hydroxyapatite co-doped with Tb 3+ and Zn 2. NANOSCALE 2023; 15:3940-3951. [PMID: 36723206 DOI: 10.1039/d2nr06795a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Hydroxyapatite (HA) is a biomimetic biomaterial that has been widely used in bone repair for many years. However, the increased risk of infection after surgery and long-time tracing for the material distribution and degradation during bone reconstruction remain challenges in the clinic. Zinc (Zn) is considered as an indispensable microelement for humans and is characterized with antibacterial action and osteogenic activity. Terbium (Tb), a rare-earth element, emits stable fluorescence under ultraviolet light. Here, Tb3+/Zn2+ co-doped hydroxyapatite (HA:Tb/Zn) was prepared to synchronously realize the antibacterial effect, osteogenic activity, and long-time tracing property. We found that HA:Tb/Zn had a strong antibacterial effect on both Gram-positive and Gram-negative clinical infectious bacteria, as well as improved osteogenic activity. HA:Tb/Zn also displayed stable green fluorescence in vitro and in vivo, which indicated great potential for recognizing the material changes during the bone reconstruction process. The combination of the ternary functions is of great significance to control the overuse of antibiotics and realize long-time tracing, and provide a versatile design on biomaterials in bone repair.
Collapse
Affiliation(s)
- Juan Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Ya-Qi Chi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yu-Jia Yan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Meng-Zhen Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Xuan Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Xue-Qin Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yuan Gao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Qin Zou
- Analytical and Testing Center, Sichuan University, Chengdu, 610064, China
| | - Ling Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Xi-Yu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
50
|
Jiang X, Liu X, Cai J, Wei S, Wang Y, Duan Z, Zhou Z, Sun R, Qu X, Tang Y. Fabrication and properties of multi-functional polydopamine coated Cu/F-codoped hydroxyapatite hollow microspheres as drug carriers. Colloids Surf B Biointerfaces 2023; 222:113097. [PMID: 36549247 DOI: 10.1016/j.colsurfb.2022.113097] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/11/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Due to its excellent bone conductivity and drug adsorption as well as pH-responsive drug release property, hydroxyapatite (HAp) is widely used as a drug carrier in bone repair field. Here, we report for the first time a novel multi-functional polydopamine (PDA) coated Cu/F-codoped HAp (Cu/F-HAp-PDA) hollow microspheres. Both Cu2+ and F- were successfully doped into the lattice of HAp and uniformly distributed in the shell of hollow microspheres through a one-step hydrothermal synthesis. Then PDA was coated homogeneously on the outer layer of Cu/F-HAp hollow microspheres. Both Cu/F-HAp and Cu/F-HAp-PDA samples displayed high drug loading efficiency and pH responsive drug release behavior. Moreover, the obtained Cu/F-HAp-PDA hollow microspheres exhibited excellent photothermal conversion efficiency and photothermal stability. The molecular dynamics simulations showed that PDA and HAp can form mutual binding mainly through Ca-O bonding, while doxorubicin (DOX) is mainly bound to PDA molecules through hydrogen bonding and π-π stacking interaction.
Collapse
Affiliation(s)
- Xiaodan Jiang
- College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiaowei Liu
- College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jiayi Cai
- College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Shibo Wei
- College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yanan Wang
- College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Zhuqing Duan
- College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Zeao Zhou
- College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Ruixue Sun
- College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Xiaofei Qu
- College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Yuanzheng Tang
- College of Electromechanical Engineering, Qingdao University of Science and Technology, Qingdao 266061, China
| |
Collapse
|