1
|
Bi X, Mao Z, Zhang Y, Ren Z, Yang K, Yu C, Chen L, Zheng R, Guan J, Liu Z, Yu B, Huang Y, Shu X, Zheng Y. Endogenous dual-responsive and self-adaptive silk fibroin-based scaffold with enhancement of immunomodulation for skull regeneration. Biomaterials 2025; 320:123261. [PMID: 40132357 DOI: 10.1016/j.biomaterials.2025.123261] [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: 12/15/2024] [Revised: 02/19/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025]
Abstract
Despite the current biomaterials (e.g. titanium mesh and polyether ether ketone) have been applied to clinical skull repair, the limitations on mechanical match, shape adaptability, bioactivity and osteointegration have greatly limited their clinical application. In this work, we constructed a water and inflammatory microenvironment dual-responsive self-adaptive silk fibroin-magnesium oxide-based scaffold with the matrix metalloproteinase-2-responsive gelatin-methacryloyl-interleukin-4 (IL-4) coating, which presented good mechanical compliance, quickly shape matching and intraoperative reprocessability. With the capability of responding to an acute inflammation microenvironment followed by a triggered on-demand release of the IL-4, the combination of immunoactive IL-4 and Mg2+ co-ordinately facilitated metabolic reprogramming by suppressing glycolysis, promoting mitochondrial oxidative phosphorylation and modulating adenosine 5'-monophosphate-activated protein kinase (AMPK) signalling pathways in macrophages, resulting in significantly facilitating M2 macrophage activation. During the stage of tissue remodelling, the sustained release of Mg2+ further promoted macrophage M2 polarization and the expression of anti-inflammatory cytokines, significantly reduced immune response and improved ectopic osteogenesis ability. Meanwhile, the cranial defect models of male rats demonstrated that this scaffold could significantly enhance biomineralized deposition and vascularisation, and achieve good bone regeneration of cranial defects. Overall, the bioactive scaffold provides a promising biomaterial and alternative repair strategy for critical-size skull defect repair.
Collapse
Affiliation(s)
- Xuewei Bi
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China; School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Zhinan Mao
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China; School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Yilin Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Zeqi Ren
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Kang Yang
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Chunhao Yu
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China; School of Life, Beijing Institute of Technology, No.5, Zhongguancun South Street, Haidian District, Beijing, China
| | - Lei Chen
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Rui Zheng
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Juan Guan
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing 100191, China
| | - Zhenhai Liu
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Binsheng Yu
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China
| | - Yongcan Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China.
| | - Xiong Shu
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
2
|
Li W, Shen Q, Tong T, Tian H, Lian X, Wang H, Yang K, Dai Z, Li Y, Chen X, Wang Q, Yang D, Wang F, Hao F, Wang L. Sequential simulation of regeneration-specific microenvironments using scaffolds loaded with nanoplatelet vesicles enhances bone regeneration. Bioact Mater 2025; 50:475-493. [PMID: 40342486 PMCID: PMC12059598 DOI: 10.1016/j.bioactmat.2025.04.018] [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: 10/12/2024] [Revised: 03/27/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
Bone regeneration is a complex and coordinated physiological process, and the different stages of this process have corresponding microenvironments to support cell development and physiological activities. However, biological scaffolds that provide different three-dimensional environments during different stages of bone regeneration are lacking. In this study, we report a novel composite scaffold (NPE@DCBM) inspired by the stages of bone regeneration; this scaffold was composed of a fibrin hydrogel loaded with nanoplatelet vesicles (NPVs), designated as NPE, and decellularized cancellous bone matrix (DCBM) microparticles. Initially, the NPE rapidly established a temporary microenvironment conducive to cell migration and angiogenesis. Subsequently, the DCBM simulated the molecular structure of bone and promoted new bone formation. In vitro, the NPVs regulated lipid metabolism in bone marrow mesenchymal stem cells (BMSCs), reprogramed the fate of BMSCs by activating the PI3K/AKT and MAPK/ERK positive feedback pathways, and increased BMSC functions, including proliferation, migration and proangiogenic potential. In vivo, NPV@DCBM accelerated bone tissue regeneration and repair. Initially, the NPE rapidly induced angiogenesis between DCBM microparticles, and subsequently, BMSCs differentiated into osteoblasts with DCBM microparticles at their core. In summary, the design of this composite scaffold that sequentially mimics different bone regeneration microenvironments may provide a promising strategy for bone regeneration, with clinical translational potential.
Collapse
Affiliation(s)
- Wenshuai Li
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- Hangzhou OrigO Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Qichen Shen
- Hangzhou OrigO Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Tong Tong
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Hongsen Tian
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Xiaowei Lian
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Haoli Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Ke Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Zhanqiu Dai
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Yijun Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xianhua Chen
- Zhejiang Institute of Medical Device Testing, Hangzhou, Zhejiang, 310016, China
| | - Qingqing Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315000, China
| | - Dan Yang
- Zhejiang DecellMatrix Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Feng Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Feng Hao
- Zhejiang DecellMatrix Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Linfeng Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| |
Collapse
|
3
|
Chu S, Li L, Zhang J, You J, Li X, Zhou Y, Huang X, Wu Q, Chen F, Bai X, Tan H, Weng J. Hierarchical interconnected porous scaffolds with regulated interfacial nanotopography exhibit antimicrobial, alleviate inflammation, neovascularization, and tissue integration for bone regeneration. Biomaterials 2025; 318:123186. [PMID: 39970602 DOI: 10.1016/j.biomaterials.2025.123186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/19/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
Novel interconnected porous scaffolds featuring suitable micro-interface structures hold significance in bone regeneration. Therefore, a hierarchical interconnected porous scaffold with nanotopography interface of pores, mimicking natural bone structure and extracellular matrix microenvironment, are designed to enhance bone regeneration by improving cell adhesion, proliferation, alleviate inflammation, and tissue integration capabilities. The scaffold is fabricated through Pickering emulsion templating method, with aminated gelatin and copper-hydroxyapatite nanoparticles serving as co-stabilizers. This process results in a dual nanoparticles-decorated interface, which could provide ample anchoring points for cells. Adjusting the ratio of the two nanoparticles leads to scaffold with different interfacial roughness. The resultant scaffold increases the number of cellular focal adhesions, enhancing cell adhesion, while its high porosity supports cell recruitment, proliferation and immunomodulation. Copper-hydroxyapatite adsorption at the pore interface reduces copper ion usage and exposes nanoparticles for direct cell contact, endowing the scaffold with enhanced antibacterial and angiogenic properties. An initial burst release phase of copper ions exerts inhibitory effects on mRNA expression, followed by a sustained and optimal release phase that promotes osteogenesis. The molecular mechanism underlying the scaffold of osteogenic potential has been elucidated through RNA sequencing analysis, along with the regulation of inflammatory cytokine expression. In vitro and in vivo studies alike verify its neovascularization-promoting capacity. The efficacy shown in a rat model with critical cranial defects underscores its clinical promise for bone regeneration, as Cu-doped scaffolds retain osteoinductive qualities after 10 weeks in vivo. This study innovates a manufacturing method for a novel scaffold in bone tissue engineering.
Collapse
Affiliation(s)
- Shirun Chu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China; Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Linlong Li
- College of Medicine (Institute of Biomedical Engineering), Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Jiahao Zhang
- College of Medicine (Institute of Biomedical Engineering), Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Jing You
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Xiaolan Li
- College of Medicine (Institute of Biomedical Engineering), Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Yuanyuan Zhou
- College of Medicine (Institute of Biomedical Engineering), Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Xiao Huang
- College of Medicine (Institute of Biomedical Engineering), Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Qiaoli Wu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Fang Chen
- Laboratory Medical Center, Jiangyou City Second People's Hospital, Mianyang 621700, Sichuan, China
| | - Xue Bai
- College of Medicine (Institute of Biomedical Engineering), Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Huan Tan
- College of Medicine (Institute of Biomedical Engineering), Southwest Jiaotong University, Chengdu 610031, Sichuan, China.
| | - Jie Weng
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China; Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China; College of Medicine (Institute of Biomedical Engineering), Southwest Jiaotong University, Chengdu 610031, Sichuan, China.
| |
Collapse
|
4
|
Liu X, Zhao Y, Wu X, Zhou Y, Liu Y, Wang S, Zhang Y, Yang H, Song F, Huang C. Spatiotemporally Programming Microenvironment to Recapitulate Endochondral Ossification via Greenhouse-Inspired Bionic Niche. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2504057. [PMID: 40317581 DOI: 10.1002/adma.202504057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/31/2025] [Indexed: 05/07/2025]
Abstract
Various biomaterials have been developed to address challenging critical-sized bone defects. However, most of them focus on intramembranous ossification (IMO) rather than endochondral ossification (ECO), often resulting in suboptimal therapeutic outcomes. Drawing inspiration from the functionality of the greenhouse ecosystem, herein a bionic niche is innovatively crafted to recapitulate the ECO process. This niche consists of three hierarchical components: an embedded microchannel network that facilitates cell infiltration and matter exchange, a polydopamine surface modification layer with immunomodulatory functions, and an ECO-targeted delivery system based on mesoporous silica nanoparticles. Through spatiotemporally programming of the microenvironment, the bionic niche effectively recapitulates the key stages of ECO. Notably, even in the rat calvaria, a region well-known for IMO, the bionic niche is capable of initiating ECO, evident by cartilage template formation, leading to efficient bone regeneration. Taken together, this study introduces prospective concepts for designing next-generation ECO-driven biomaterials for bone tissue engineering.
Collapse
Affiliation(s)
- Xuzheng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yaning 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, 430079, China
| | - Xiaoyi Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yueli Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yingheng Liu
- Dental Materials Science, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Shilei Wang
- Key Laboratory of Resources and Compound of Traditional Chinese Medicine, Ministry of Education, Hubei University of Traditional Chinese Medicine, Wuhan, 430065, China
| | - 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, 430079, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
5
|
Cao B, Zhang K, Zuo R, Kang Z, Lin J, Kang Z, Luo D, Chai Y, Xu J, Kang Q, Qiu S. 3D-bioprinted functional scaffold based on synergistic induction of i-PRF and laponite exerts efficient and personalized bone regeneration via miRNA-mediated TGF-β/Smads signaling. Int J Surg 2025; 111:3193-3211. [PMID: 40035712 DOI: 10.1097/js9.0000000000002312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 02/17/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Limited stem cells, low vascularization efficiency, and weak osteoinductive activity plague the repair and reconstruction of bone defects with cell-free scaffolds. METHODS Herein, injectable platelet-rich fibrin (i-PRF) was loaded into a alginate methacryloyl (AlgMA)/gelatin methacryloyl (GelMA)-methylcellulose (AGM) bioink system and constructed a porous hydrogel scaffold by 3D bioprinting. The addition of nanosilicate-laponite (Lap) further enhanced this scaffold and synergized with i-PRF to promote efficient and personalized cranial regeneration. RESULTS At the biochemical level, Lap significantly enhanced the ability of the scaffold to retard growth factor release, and multiple physiologically proportional growth factors in the scaffold synergistically promoted rapid neoangiogenesis and concomitantly recruited endogenous bone marrow mesenchymal stem cells (BMSCs). More importantly, the bioactive ions released by Lap markedly promoted the proliferation of BMSCs and consistently induced the osteogenic differentiation of BMSCs. At the immunological level, i-PRF-AGM@Lap significantly attenuates the inflammatory response by promoting macrophage M2 polarization. Mechanistically, miRNA sequencing and functional validation experiments demonstrated that bioactive ions released by Lap could synergize with growth factors in i-PRF to promote osteogenic differentiation of BMSCs through the miR-21 and miR-125a-mediated transforming growth factor-β/Smads signaling pathway. CONCLUSION The results of this study provide a new idea for the personalized treatment of bone defects.
Collapse
Affiliation(s)
- Bojun Cao
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical and Translational Research Center for 3D Printing Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kunqi Zhang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongtai Zuo
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyang Kang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieming Lin
- Department of Orthopaedic Surgery, Renji Hospital, South Campus, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhixuan Kang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dinghao Luo
- Clinical and Translational Research Center for 3D Printing Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimin Chai
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Xu
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qinglin Kang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Qiu
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Xiao Y, Qu Y, Hu X, Zhao J, Xu S, Zheng L, Liang X. E7 peptide modified poly(ε-caprolactone)/silk fibroin/octacalcium phosphate nanofiber membranes with "recruitment-osteoinduction" potentials for effective guided bone regeneration. Int J Biol Macromol 2025; 305:140862. [PMID: 39952537 DOI: 10.1016/j.ijbiomac.2025.140862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Effective membranes that have the osteogneic potential and the ability to recruit osteoblast precursor cells challenged guided bone regeneration (GBR). Herein, we engineered multifunctional nanofiber membranes by using eletrospun poly(ε-caprolactone) (PCL) and silk fibroin (SF), incorporated with octacalcium phosphate (OCP) and BMSCs-affine peptide (E7) to form the PCL/SF/OCP/E7 (PSOE) nanofiber, wherein the E7 peptide enhances the enrichment of BMSCs, and OCP as osteogenesis promoter. The composite membranes enhance the recruitment and biomineralization processes essential for bone regeneration. Notably, the dual functionality of BMSC recruitment and osteoinduction provides a "recruitment-osteoinduction" strategy that significantly improves bone repair. In vitro analyses confirmed that the PSOE nanofibers have superior hydrophilicity and biocompatibility, and significantly upregulated the expression of osteogenic genes in mesenchymal stem cells, thereby facilitating osteogenic differentiation. In vivo studies using a rat tibial defect model revealed that PSOE nanofibers promoted bone repair within 8 weeks, as validated by micro-CT and histological evaluations. This study highlights the PSOE nanofiber's potential as a promising synthetic periosteum substitute for effective bone regeneration.
Collapse
Affiliation(s)
- Yuanming Xiao
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China; Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, International Joint Laboratory on Regeneration of Bone and Soft Tissues, Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, PR China
| | - Yangyang Qu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, International Joint Laboratory on Regeneration of Bone and Soft Tissues, Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, PR China
| | - Xuankai Hu
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China; Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, International Joint Laboratory on Regeneration of Bone and Soft Tissues, Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, PR China
| | - Jinmin Zhao
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China; Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, International Joint Laboratory on Regeneration of Bone and Soft Tissues, Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, PR China
| | - Sheng Xu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, International Joint Laboratory on Regeneration of Bone and Soft Tissues, Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, PR China; Life Science Research Institute of Guangxi Medical University, Nanning, 530021, PR China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, International Joint Laboratory on Regeneration of Bone and Soft Tissues, Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, PR China.
| | - Xiaonan Liang
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China.
| |
Collapse
|
7
|
Liu H, Wei J, Xiao S, Jin S, Yuan L, Wen J, Liu J, Li Y, Li J. Regulation Energy Metabolism of Fiber Scaffolds Orchestrates Osteoimmunomodulation and Angio/Osteogenesis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409747. [PMID: 40135330 DOI: 10.1002/smll.202409747] [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: 11/06/2024] [Indexed: 03/27/2025]
Abstract
Bone formation is a highly metabolic process, involving extensive biosynthesis and biomineralization, both of which require substantial amounts of energy. Additionally, the regulation of the immune microenvironment and the development of a neovascularization network are equally crucial in bone formation. Inspired by the high energy demands of the bone formation process, a core-shell electrospun fiber scaffold (PFC/PCK) capable of sustainably releasing a metabolic regulator (αKG) and biomineralizing ions (CaP) is developed. In vitro experiments show that the PFC/PCK fiber scaffolds can induce hyperpolarization of mitochondrial membrane potential in bone marrow mesenchymal stem cells (BMSCs), increase energy supply, effectively regulate immune microenvironment, and remarkably promote expression of angiogenesis and osteogenesis markers. In vivo evaluation further confirms the outstanding immunoregulatory and osteo/angio-genesis capabilities of the fabricated fiber scaffolds. Importantly, transcriptome analysis identifies that the fiber scaffolds upregulate genes and signaling pathways associated with M2 macrophage activation, energy generation, angiogenesis, and osteogenesis. Additionally, metabolomics analysis confirms that the fiber scaffolds enhance bone formation by promoting the expression of osteogenic metabolites. The versatile fiber scaffolds developed in current study demonstrates a new strategy for functional bone regeneration.
Collapse
Affiliation(s)
- Huan Liu
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, China
- North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Jiawei Wei
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, China
| | - Shiqi Xiao
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, China
| | - Shue Jin
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, China
| | - Li Yuan
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, China
| | - Jing Wen
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, China
| | - Jiangshan Liu
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, China
| | - Yubao Li
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, China
| | - Jidong Li
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
8
|
Hang R, Yao X, Bai L, Hang R. Evolving biomaterials design from trial and error to intelligent innovation. Acta Biomater 2025; 197:29-47. [PMID: 40081552 DOI: 10.1016/j.actbio.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/20/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The design and exploration of biomaterials plays a pivotal role in many fields, including medical and engineering. The prevailing approach to biomaterials discovery relies on orthogonal experiments, with repeated attempts to optimize experimental conditions. This method has proven invaluable in gaining experience, but it is also inefficient and challenging to predict the behavior of complex systems. The advent of high-throughput screening (HTS) techniques has led to a notable enhancement in the efficiency of biomaterials development, enabling researchers to assess a vast array of material combinations within a relatively short timeframe. Nevertheless, the emergence of artificial intelligence (AI) has been the catalyst for a new era in biomaterials design. AI has markedly accelerated the development of new materials by enabling the prediction of material properties through machine learning (ML) and deep learning models, as well as optimizing the design pipeline. This review will present a systematic overview of the development of biomaterials design technology. It will also explore the integration of AI with HTS technology and envisage the potential of AI-driven materials design in biomaterials for the future. STATEMENT OF SIGNIFICANCE: The design and synthesis of biomaterials have undergone substantial shifts, reflecting evolving research paradigms. High-throughput screening has emerged as a broad and efficient alternative to traditional free-form combination methods in biomaterial design. The advent of artificial intelligence (AI) enables personalized biomaterial design and, as a transformative tool in biomaterial development, is poised to redefine the field and offer long-term solutions for its advancement. Building on these advancements, this review systematically summarizes the evolution of biomaterial design, offering insights into the future trajectory of the field.
Collapse
Affiliation(s)
- Ruiyue Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, PR China.
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China.
| |
Collapse
|
9
|
Khorasani E, Vahidi B. 3D-Printed Scaffolds for Cranial Bone Regeneration: A Systematic Review of Design, Materials, and Computational Optimization. Biotechnol Bioeng 2025. [PMID: 40289530 DOI: 10.1002/bit.28994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/26/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025]
Abstract
Cranial bone defects from trauma, congenital conditions, or surgery are challenging to treat due to the skull's limited regeneration. Traditional methods like autografts and allografts have drawbacks, including donor site issues and poor integration. 3D-printed scaffolds provide a patient-specific alternative, improving bone regeneration and integration. This review evaluates advancements in 3D-printed scaffolds for cranial bone regeneration, focusing on fabrication techniques, material innovations, and structural optimization while assessing their preclinical and clinical potential. A systematic literature search (2014-2024) was conducted using PubMed and other databases. Studies addressing scaffold properties such as porosity, pore interconnectivity, and mechanical stability were included, while non-cranial scaffold studies were excluded. Advances in 3D printing have enabled patient-specific scaffolds with optimized architecture to enhance bone regeneration, mechanical support, and nutrient transport. Bioceramics, polymers, and composites mimic native bone properties, while bioactive coatings further improve osteogenesis. However, limited clinical translation and insufficient customization remain challenges. Further preclinical and clinical trials are crucial to overcoming barriers in mechanical optimization and patient-specific scaffold fabrication, bridging the gap between research and clinical applications.
Collapse
Affiliation(s)
- Elnaz Khorasani
- Department of Medical Technology and Tissue Engineering, Faculty of Life Science Engineering, School of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran
| | - Bahman Vahidi
- Department of Medical Technology and Tissue Engineering, Faculty of Life Science Engineering, School of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran
| |
Collapse
|
10
|
Wang H, Zhang J, Li Z, Liu J, Chang H, Jia S, Di Z, Liu H, Wang J, Gao D, Wang C, Li G, Zhao X. NIR-programmable 3D-printed shape-memory scaffold with dual-thermal responsiveness for precision bone regeneration and bone tumor management. J Nanobiotechnology 2025; 23:300. [PMID: 40247322 PMCID: PMC12007331 DOI: 10.1186/s12951-025-03375-1] [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: 12/07/2024] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
Clinically, intraoperative treatment of bone tumors presents several challenges, including the effective inactivation of tumors and filling of irregular bone defects after tumor removal. In this study, intelligent thermosensitive composite materials with shape-memory properties were constructed using polylactic acid (PLA) and polycaprolactone (PCL), which have excellent biocompatibility and degradability. Additionally, beta-tricalcium phosphate (β-TCP), with its osteogenic properties, and magnesium (Mg) powder, with its photothermal and bone-promoting abilities, were incorporated to improve the osteogenic potential of the composite and enable the material to respond intelligently to near-infrared (NIR) light. Utilizing 3D printing technology, the composite material was prepared into an NIR-responsive shape-memory bone-filling implant that deforms when the scaffold temperature increases to 48 ℃ under NIR laser irradiation. Moreover, at a lower temperature of 42 ℃, mild photothermal therapy promotes macrophage polarization toward the M2 phenotype. This process regulates the secretion of interleukin (IL)-4, IL-10, tumor necrosis factor-α, IL-6, and bone morphogenetic protein (BMP)-2, reducing local inflammation, enhancing the release of pro-healing factors, and improving osteogenesis. Overall, this innovative scaffold is a promising and efficient treatment for filling irregular bone defects after bone tumor surgery.
Collapse
Affiliation(s)
- Hui Wang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Jiaxin Zhang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zuhao Li
- Department of Orthopaedics, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Jiaqi Liu
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Haoran Chang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Shipu Jia
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zexin Di
- Department of Orthopaedics, School of Economics and Management, Beihua University, Jilin, 132013, China
| | - He Liu
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Jincheng Wang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Delong Gao
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, 130022, China.
| | - Chenyu Wang
- Department of Plastic & Reconstruct Surgery, First Hospital of Jilin University, Changchun, 130061, China.
| | - Guiwei Li
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun, 130025, China.
| | - Xin Zhao
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
11
|
Xue B, Xu Z, Li L, Guo K, Mi J, Wu H, Li Y, Xie C, Jin J, Xu J, Jiang C, Gu X, Qin M, Jiang Q, Cao Y, Wang W. Hydrogels with programmed spatiotemporal mechanical cues for stem cell-assisted bone regeneration. Nat Commun 2025; 16:3633. [PMID: 40240370 PMCID: PMC12003706 DOI: 10.1038/s41467-025-59016-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
Hydrogels are extensively utilized in stem cell-based tissue regeneration, providing a supportive environment that facilitates cell survival, differentiation, and integration with surrounding tissues. However, designing hydrogels for regenerating hard tissues like bone presents significant challenges. Here, we introduce macroporous hydrogels with spatiotemporally programmed mechanical properties for stem cell-driven bone regeneration. Using liquid-liquid phase separation and interfacial supramolecular self-assembly of protein fibres, the macroporous structure of hydrogels provide ample space to prevent contact inhibition during proliferation. The rigid protein fibre-coated pore shell provides sustained mechanical cues for guiding osteodifferentiation and protecting against mechanical loads. Temporally, the hydrogel exhibits tunable degradation rates that can synchronize with new tissue deposition to some extent. By integrating localized mechanical heterogeneity, macroporous structures, surface chemistry, and regenerative degradability, we demonstrate the efficacy of these stem cell-encapsulated hydrogels in rabbit and porcine models. This marks a substantial advancement in tailoring the mechanical properties of hydrogels for stem cell-assisted tissue regeneration.
Collapse
Affiliation(s)
- Bin Xue
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| | - Zhengyu Xu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), MOE Key Laboratory of High Performance Polymer Materials and Technology, and State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China
| | - Kaiqiang Guo
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Jing Mi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Haipeng Wu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Yiran Li
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Chunmei Xie
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jing Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Juan Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chunping Jiang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Xiaosong Gu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Meng Qin
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China.
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center (ChemBIC), MOE Key Laboratory of High Performance Polymer Materials and Technology, and State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
12
|
Yan Z, Deng Y, Huang L, Zeng J, Wang D, Tong Z, Fan Q, Tan W, Yan J, Zang X, Chen S. Biopolymer-based bone scaffold for controlled Pt (IV) prodrug release and synergistic photothermal-chemotherapy and immunotherapy in osteosarcoma. J Nanobiotechnology 2025; 23:286. [PMID: 40205459 PMCID: PMC11983740 DOI: 10.1186/s12951-025-03253-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025] Open
Abstract
Achieving bone defect repair while preventing tumor recurrence after osteosarcoma surgery has consistently posed a clinical challenge. Local treatment with 3D-printed scaffolds loaded with chemotherapeutic drugs can exert certain effects in tumor inhibition and bone regeneration. However, the non-specific activation of chemotherapeutic drugs leads to high local toxic side effects and the formation of an immunosuppressive tumor microenvironment, thereby limiting their clinical application and therapeutic efficacy. To address this, we designed a Pt (IV) prodrug with low toxicity and minimal side effects, which releases Pt (II) in response to glutathione. This prodrug was grafted onto polydopamine (PDA) through an amidation reaction, resulting in a composite nanomaterial (PDA@Pt) that possesses both photothermal synergistic chemotherapy and immuno-oncological properties. Subsequently, we innovatively employed selective laser sintering technology to incorporate PDA@Pt into a poly (L-lactic acid)/bioactive glass matrix, successfully constructing a composite scaffold with dual anti-tumor and bone repair capabilities. The study revealed that the composite scaffold significantly inhibited the growth of osteosarcoma cells and activated the cGAS-STING pathway by inducing DNA damage, ultimately converting the 'cold tumor' into a 'hot tumor.' Additionally, the composite scaffold could induce osteogenic differentiation of bone marrow mesenchymal stem cells and exhibited excellent bone repair capabilities in vivo.
Collapse
Affiliation(s)
- Zuyun Yan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Liping Huang
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jin Zeng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Dong Wang
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhaochen Tong
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Qizhi Fan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Wei Tan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jinpeng Yan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, 410017, P. R. China
| | - Xiaofang Zang
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Shijie Chen
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China.
| |
Collapse
|
13
|
Miao S, Guo J, Zhang Y, Liu P, Chen X, Han Q, Wang Y, Xuan K, Yang P, Tao F. Biomimetic Intrafibrillar Mineralization of Hierarchically Structured Amyloid-Like Fibrils. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2416824. [PMID: 40195686 DOI: 10.1002/adma.202416824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/26/2025] [Indexed: 04/09/2025]
Abstract
Intrafibrillar mineralization is essential not only as a fundamental process in forming biological hard tissues but also as a foundation for developing advanced composite fibril-based materials for innovative applications. Traditionally, only natural collagen fibrils have been shown to enable intrafibrillar mineralization, presenting a challenge in designing ordered hierarchical fibrils from common protein aggregation that exhibit similar high intrafibrillar mineralization activity. In this study, a mechanically directed two-step transformation method is developed that converts phase-transitioned protein nanofilms into crystalline, hierarchical amyloid-like fibrils with multilayer structures, which effectively control the growth and lateral organization of hydroxyapatite within adaptive gaps. The resulting mineralized HSAF achieves a hardness of 0.616 ± 0.007 GPa and a modulus of 19.06 ± 3.54 GPa-properties closely resembling native hard tissues-and exhibits exceptionally high bioactivity in promoting both native bone tissue growth and further intrafibrillar mineralization, achieving 76.9% repair in a mice cranial defect model after 8 weeks and outperforming other regenerative materials. This remarkable performance, stemming from the unique structure and composition of the fibers, positions HSAF as a promising candidate for biomedical and engineering applications. These findings advance the understanding of biomineralization mechanisms and establish a foundation for developing high-bioactivity materials for hard tissue regeneration.
Collapse
Affiliation(s)
- Shuting Miao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Jing Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yuexin Zhang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Peisheng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaojie Chen
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Qian Han
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Yingbo Wang
- College of Chemical Engineering, Xinjiang Key Laboratory of Energy Storage and Photoelectroctalytic Materials, Xinjiang Normal University, 102 Xinyi Road, Urumqi, 830054, China
| | - Kun Xuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Fei Tao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| |
Collapse
|
14
|
Han J, Li Z, Du J, Zhang Q, Ge S, Liu H, Ma B. Natural collagen scaffold with intrinsic piezoelectricity for enhanced bone regeneration. Mater Today Bio 2025; 31:101532. [PMID: 39968523 PMCID: PMC11834078 DOI: 10.1016/j.mtbio.2025.101532] [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: 11/25/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Materials-mediated piezoelectric signals have been widely applied in bone regeneration. Collagen is the most abundant protein in the human body, and native collagen with complete tertiary structure shows efficient piezoelectricity. However, the traditional collagen scaffolds are lack of piezoelectricity due to the destruction of the complete tertiary structure. Here, natural collagen scaffolds with the complete tertiary structure were prepared. Alkali treatment made the collagen scaffold lose piezoelectricity. The collagen with/without piezoelectricity (PiezoCol/NCol) scaffolds both possessed good cytocompatibility and promoted cell adhesion. After being implanted subcutaneously, the NCol scaffold almost did not affect bone regeneration with/without ultrasound treatment. However, under ultrasound treatment, the PiezoCol scaffold promoted the new bone formation with enhanced osteogenic differentiation, angiogenesis, and neural differentiation, meaning that piezoelectricity endows collagen with satisfactory promotion for bone regeneration. Meanwhile, the PiezoCol scaffold can also accelerate bone formation without ultrasound treatment, which should be attributed to the daily exercise-caused weak piezoelectric stimulation. Further, the proteomic analysis revealed the mechanism by which the PiezoCol scaffold promoted bone tissue formation via mainly upregulating the PI3K-Akt signaling pathway. This study provides a new strategy to enhance the osteoinduction of collagen scaffold for bone regeneration by maintaining intrinsic piezoelectricity.
Collapse
Affiliation(s)
- Jing Han
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| | - Zhao Li
- College of Materials Science and Engineering, Qingdao University of Science & Technology, Qingdao, Shandong, 266061, China
| | - Jing Du
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| | - Qun Zhang
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| | - Shaohua Ge
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250013, China
| | - Baojin Ma
- Department of Tissue Engineering & Periodontology, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, China
| |
Collapse
|
15
|
Li B, Ma Y, Fatima K, Zhou X, Gu X, Chen S, He C. 3D printed shape-memory piezoelectric scaffolds with in-situ self-power properties for bone defect repair. J Nanobiotechnology 2025; 23:244. [PMID: 40128753 PMCID: PMC11934793 DOI: 10.1186/s12951-025-03325-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/13/2025] [Indexed: 03/26/2025] Open
Abstract
Electrical stimulation has been shown to regulate early immunity and late-stage osteogenesis in bone repair. However, achieving in-situ electrical stimulation in the form of self-power in vivo during the initial postoperative stages when the patients have limited mobility remains challenging. In this study, we developed a 3D-printed in-situ self-powered composite scaffold composed of shape memory polyurethane elastomers (SMPU) and polyvinylidene fluoride (PVDF) piezoelectric nanofibers. The composite scaffold demonstrates excellent shape memory performance, allowing for minimally invasive implantation. During the shape memory process, the composite scaffold can provide mechanical force stimulation to PVDF nanofibers to generate charge. Therefore, self-power was achieved through the integration of the shape memory process and piezoelectric effects, and it can be used for in-situ electrical stimulation during the initial postoperative period. Additionally, the composite scaffold can output voltage under continuous mechanical force stimulation, indicating that the patients can apply sustained mechanical force stimulation to the composite scaffold to output voltage through rehabilitation exercises when the patients regain mobility. Both cell experiments and animal studies confirmed that this composite scaffold can effectively regulate the immune microenvironment and enhance osteogenesis. This study successfully achieves in-situ electrical stimulation in the form of self-power by integrating the shape memory process and piezoelectric effects, which is expected to be an effective repair strategy for bone tissue engineering.
Collapse
Affiliation(s)
- Bing Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, People's Republic of China
| | - Yichao Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Kanwal Fatima
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, People's Republic of China
| | - Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, People's Republic of China
| | - Xin Gu
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, People's Republic of China.
| | - Shuo Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, People's Republic of China.
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China.
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, People's Republic of China.
| |
Collapse
|
16
|
Li B, Ma Y, Fatima K, Zhou X, Chen S, He C. 3D printed scaffolds with multistage osteogenic activity for bone defect repair. Regen Biomater 2025; 12:rbaf010. [PMID: 40151200 PMCID: PMC11947418 DOI: 10.1093/rb/rbaf010] [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: 11/27/2024] [Revised: 01/20/2025] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
The bone defect repair is a complex process including immune regulation, stem cell osteogenic differentiation and extracellular matrix mineralization. Current bone tissue engineering approaches often fail to adapt throughout the above osteogenic process, resulting in suboptimal repair outcomes. To address this problem, a 3D-printed scaffold with multistage osteogenic activity based on shape-memory elastomer and electroactive material is developed. The scaffold exhibits excellent shape memory performance and can trigger shape recovery by physiological temperature. The physiological temperature-triggered shape-memory behavior makes the scaffold promising for minimally invasive implantation. After electric field polarization, the scaffold's surface carries the negative charge, which can activate the PI3K/Akt signaling pathway to promote the polarization of macrophages to M2 phenotype and activate the FAK/ERK signaling pathway to promote osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), indicating that the scaffold can effectively participate in immune microenvironment regulation and stem cell osteogenic differentiation. Additionally, the negative charge on the scaffold's surface can attract calcium and phosphate ions, forming a mineralized matrix and promoting late-stage extracellular matrix mineralization by continuously supplying mineralizing ions such as calcium and phosphate. Overall, this study introduces a 3D-printed scaffold with multistage osteogenic activity, offering a promising strategy for bone defect repair.
Collapse
Affiliation(s)
- Bing Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yichao Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Kanwal Fatima
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Shuo Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
17
|
Li X, Tang J, Guo W, Dong X, Cao K, Tang F. Polydopamine Nanocomposite Hydrogel for Drug Slow-Release in Bone Defect Repair: A Review of Research Advances. Gels 2025; 11:190. [PMID: 40136895 PMCID: PMC11942372 DOI: 10.3390/gels11030190] [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: 02/16/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
In recent years, hydrogels have emerged as promising candidates for bone defect repair due to their excellent biocompatibility, high porosity, and water-retentive properties. However, conventional hydrogels face significant challenges in clinical translation, including brittleness, low mechanical strength, and poorly controlled drug degradation rates. To address these limitations, as a multifunctional polymer, polydopamine (PDA) has shown great potential in both bone regeneration and drug delivery systems. Its robust adhesive properties, biocompatibility, and responsiveness to photothermal stimulation make it an ideal candidate for enhancing hydrogel performance. Integrating PDA into conventional hydrogels not only improves their mechanical properties but also creates an environment conducive to cell adhesion, proliferation, and differentiation, thereby promoting bone defect repair. Moreover, PDA facilitates controlled drug release, offering a promising approach to optimizing treatment outcomes. This paper first explores the mechanisms through which PDA promotes bone regeneration, laying the foundation for its clinical translation. Additionally, it discusses the application of PDA-based nanocomposite hydrogels as advanced drug delivery systems for bone defect repair, providing valuable insights for both research and clinical translation.
Collapse
Affiliation(s)
- Xiaoman Li
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China; (X.L.); (W.G.); (X.D.); (K.C.)
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi 563006, China
| | - Jianhua Tang
- Cancer Research UK Manchester Institute, The University of Manchester, Cheshire SK10 4TG, UK;
| | - Weiwei Guo
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China; (X.L.); (W.G.); (X.D.); (K.C.)
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi 563006, China
| | - Xuan Dong
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China; (X.L.); (W.G.); (X.D.); (K.C.)
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi 563006, China
| | - Kaisen Cao
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China; (X.L.); (W.G.); (X.D.); (K.C.)
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi 563006, China
| | - Fushan Tang
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China; (X.L.); (W.G.); (X.D.); (K.C.)
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi 563006, China
| |
Collapse
|
18
|
Zhao J, Sarkar N, Ren Y, Pathak AP, Grayson WL. Engineering next-generation oxygen-generating scaffolds to enhance bone regeneration. Trends Biotechnol 2025; 43:540-554. [PMID: 39343620 PMCID: PMC11867879 DOI: 10.1016/j.tibtech.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/08/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
In bone, an adequate oxygen (O2) supply is crucial during development, homeostasis, and healing. Oxygen-generating scaffolds (OGS) have demonstrated significant potential to enhance bone regeneration. However, the complexity of O2 delivery and signaling in vivo makes it challenging to tailor the design of OGS to precisely meet this biological requirement. We review recent advances in OGS and analyze persisting engineering and translational hurdles. We also discuss the potential of computational and machine learning (ML) models to facilitate the integration of novel imaging data with biological readouts and advanced biomanufacturing technologies. By elucidating how to tackle current challenges using cutting-edge technologies, we provide insights for transitioning from traditional to next-generation OGS to improve bone regeneration in patients.
Collapse
Affiliation(s)
- Jingtong Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Naboneeta Sarkar
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Yunke Ren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Arvind P Pathak
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA; Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
19
|
Wu X, Yang H, Liu G, Sun W, Li J, Zhao Y, Gao X, Liu X, Song F, Wang S, Lu Z, Chen C, Huang C. Osteomimix: A Multidimensional Biomimetic Cascade Strategy for Bone Defect Repair. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416715. [PMID: 39924776 DOI: 10.1002/adma.202416715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/24/2025] [Indexed: 02/11/2025]
Abstract
Despite advancements in biomimetic mineralization techniques, the repair of large-scale bone defects remains a significant challenge. Inspired by the bone formation process, a multidimensional biomimetic cascade strategy is developed by replicating the biomineralization cascade, emulating the hierarchical structure of bone, and biomimicking its biological functions for efficient bone regeneration. This strategy involves the photocrosslinking of sodium methacrylate carboxymethyl cellulose-stabilized amorphous magnesium-calcium phosphate with methacrylate-modified type I collagen to create a self-mineralizing hydrogel. The hydrogel is then integrated with either naturally derived or synthetic oriented bulk scaffolds. The resulting composite, named Osteomimix, provides excellent mechanical support and can be customized for irregular bone defects using CAD/CAM technology. Through in vitro and in vivo studies, this work finds that Osteomimix exhibits spontaneous in situ biomimetic mineralization in a cell-free environment, while modulating immune responses and promoting vascularized bone formation in a cell-dependent manner. Built on bone-specific insights, this strategy achieves biomimicry across temporal, spatial, and functional dimensions, facilitating the seamless integration of artificial constructs with the natural tissue repair dynamics.
Collapse
Affiliation(s)
- Xiaoyi Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, School of Resource and Environmental Sciences, Wuhan University, Wuhan, 430079, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Gufeng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Wei Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jiyun Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yaning Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xin Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xuzheng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Shilei Wang
- Hubei Engineering Center of Natural Polymer-Based Medical Materials and Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Ziyang Lu
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, School of Resource and Environmental Sciences, Wuhan University, Wuhan, 430079, China
| | - Chaoji Chen
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, School of Resource and Environmental Sciences, Wuhan University, Wuhan, 430079, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
20
|
Lam W, Yao Y, Tang C, Wang Y, Yuan Q, Peng L. Bifunctional mesoporous HMUiO-66-NH 2 nanoparticles for bone remodeling and ROS scavenging in periodontitis therapy. Biomaterials 2025; 314:122872. [PMID: 39383779 DOI: 10.1016/j.biomaterials.2024.122872] [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: 04/08/2024] [Revised: 09/17/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Periodontal bone defects represent an irreversible consequence of periodontitis associated with reactive oxygen species (ROS). However, indiscriminate removal of ROS proves to be counterproductive for tissue repair and insufficient for addressing existing bone defects. In the treatment of periodontitis, it is crucial to rationally alleviate local ROS while simultaneously promoting bone regeneration. In this study, Zr-based large-pore hierarchical mesoporous metal-organic framework (MOF) nanoparticles (NPs) HMUiO-66-NH2 were successfully proposed as bifunctional nanomaterials for bone regeneration and ROS scavenging in periodontitis therapy. HMUiO-66-NH2 NPs demonstrated outstanding biocompatibility both in vitro and in vivo. Significantly, these NPs enhanced the osteogenic differentiation of bone mesenchymal stem cells (BMSCs) under normal and high ROS conditions, upregulating osteogenic gene expression and mitigating oxidative stress. Furthermore, in vivo imaging revealed a gradual degradation of HMUiO-66-NH2 NPs in periodontal tissues. Local injection of HMUiO-66-NH2 effectively reduced bone defects and ROS levels in periodontitis-induced C57BL/6 mice. RNA sequencing highlighted that differentially expressed genes (DEGs) are predominantly involved in bone tissue development, with notable upregulation in Wnt and TGF-β signaling pathways. In conclusion, HMUiO-66-NH2 exhibits dual functionality in alleviating oxidative stress and promoting bone repair, positioning it as an effective strategy against bone resorption in oxidative stress-related periodontitis.
Collapse
Affiliation(s)
- Waishan Lam
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yufei Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, China
| | - Chenxi Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yue Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Surgery, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lin Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
21
|
Lei H, Cao H, Chen X, Su Z, Deng S, Hu Y, Wu L, Gui X, Gao C, Jia X, Pei X, Tan Z, Yuan T, Wang Q, Zhou C, Fan Y, Zhang X. A Functionalized 3D-Printed Ti6Al4V "Cell Climbing Frame" Inspired by Marine Sponges to Recruit and Rejuvenate Autologous BMSCs in Osteoporotic Bone Repair. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413238. [PMID: 39910833 DOI: 10.1002/adma.202413238] [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: 09/04/2024] [Revised: 01/09/2025] [Indexed: 02/07/2025]
Abstract
Osteoporosis, characterized by low bone mass and high fracture risk, challenges orthopedic implant design. Conventional 3D-printed Ti6Al4V scaffolds are mechanically robust but suffer from poor bone regeneration in osteoporotic patients due to stress shielding and cellular senescence. In this study, a functionalized 3D-printed Ti6Al4V "Cell Climbing Frame" is developed, aiming to adapt to the mechanical microenvironment of osteoporosis, effectively recruit and support the adhesion and growth of autologous bone marrow mesenchymal stem cells (BMSCs), while rejuvenating senescent cells for improved bone regeneration. Inspired by marine sponges, the processing accuracy limitations of selective laser melting (SLM) technology is broke through innovatively constructing a hierarchical porous structure with macropores and micropores nested within each other. Results demonstrate that the unique hierarchical porous scaffold reduces the elastic modulus, facilitates blood penetration, and enhances cell adhesion and growth. Further surface functionalization with E7 peptides and exosomes promotes the attraction and rejuvenation of BMSCs and boosts migration, proliferation, and osteogenic differentiation in vitro. In vivo, the functionalized "Cell Climbing Frame" accelerates bone repair in osteoporotic rats, while delaying surrounding bone loss, enabling robust multi-stage osseointegration. This innovation advances 3D-printed regenerative implants for osteoporotic bone repair.
Collapse
Affiliation(s)
- Haoyuan Lei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Hongfu Cao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xi Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Zixuan Su
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Siyan Deng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuxin Hu
- School of Mechanical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Lina Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xingyu Gui
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Canyu Gao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xibiao Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xuan Pei
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhen Tan
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Tun Yuan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Sichuan Testing Center for Biomaterials and Medical Devices Co.Ltd, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| |
Collapse
|
22
|
Liu J, Wei J, Xiao S, Yuan L, Liu H, Zuo Y, Li Y, Li J. Multienzyme-Activity Sulfur Quantum Dot Nanozyme-Mediated Cascade Reactions in Whole-Stage Symptomatic Therapy of Infected Bone Defects. ACS NANO 2025; 19:6858-6875. [PMID: 39936642 DOI: 10.1021/acsnano.4c12343] [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: 02/13/2025]
Abstract
Integrating the therapeutic efficacy of early bacterial clearance, midstage inflammatory remission, and late-stage effective tissue healing is considered a pivotal challenge in symptomatic treatment of infected bone defects (IBDs). Herein, a microenvironment-adaptive nanoplatform based on a sulfur quantum dot (SQD) nanozyme was proposed for whole-stage symptomatic therapy of IBDs by mediating the sequence of enzyme cascade reactions. The SQD nanozyme prepared by a size-engineering modification strategy exhibits enhanced multienzyme activity compared to conventional micrometer- and nanometer-sized sulfur particles. In the early stages of bacterial infection, the SQD nanozyme self-activates superoxide dismutase-peroxidase activity, resulting in the production of reactive oxygen species (ROS) that effectively eliminate bacteria. After disinfection, the SQD nanozyme self-switched to superoxide dismutase-catalase mimetic behavior and eliminated excess ROS, efficiently promoting macrophage polarization to an anti-inflammatory phenotype in the midinflammatory microenvironment. Importantly, SQD nanozyme-mediated M2 macrophage polarization significantly improved the damaged bone immune microenvironment, accelerating bone repair at late-stage tissue healing. Therefore, this strategy offers a promising and viable approach for the treatment of infectious tissue healing by developing multienzyme-activity nanozymes that respond intelligently to the microenvironment at different stages, effectively fighting bacteria, reducing inflammation, and promoting tissue regeneration for whole-stage symptomatic therapy.
Collapse
Affiliation(s)
- Jiangshan Liu
- Research Center for Nano-Biomaterial, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Jiawei Wei
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Shiqi Xiao
- Clinical Medical College & Affiliated Hospital, Chengdu University, Chengdu 610081, China
| | - Li Yuan
- Research Center for Nano-Biomaterial, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Huan Liu
- Research Center for Nano-Biomaterial, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Yi Zuo
- Research Center for Nano-Biomaterial, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Yubao Li
- Research Center for Nano-Biomaterial, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Jidong Li
- Research Center for Nano-Biomaterial, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| |
Collapse
|
23
|
Hou Q, He X, Guo M, Li X, Zhang Z, Xu X, Xu Y, Shi Q, Han Y. Enhanced hemostatic efficacy of cryogel with copper ion-loaded mesoporous bioactive glasses for acute and persistent bleeding. J Nanobiotechnology 2025; 23:102. [PMID: 39939976 PMCID: PMC11823261 DOI: 10.1186/s12951-025-03142-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/19/2025] [Indexed: 02/14/2025] Open
Abstract
Uncontrolled acute and persistent bleeding, as well as with infection, is a great challenge because of the high mortality during treating the patients with injuries, complex surgery or bone marrow failure. Here, we develop an external form of natural components which is based on phosphorylated methacrylated gelatin (GelMA, G) cryogel (GP) loaded with tannic acid (TA)-mixed copper ion (Cu2+) mesoporous bioactive glasses (MBG), named after GP@MBG-Cu-TA cryogel, to address the goals of reduce persistent bleeding and enhance antibacterial activity. Structurally, GP@MBG-Cu-TA cryogel is based on GP, MBG loaded with TA and Cu2+ adheres to GP via hydrogen bonding. In vitro, GP@MBG-Cu-TA cryogel displays a good biocompatibility, hemostatic and antimicrobial capability. In vivo studies, GP@MBG-Cu-TA cryogel can enhance the hemostatic effect in the liver injury in SD rats for the acute bleeding, as well as in the aplastic anemia and hemophilia A mice with tail amputation for the persistent bleeding. In addition, GP@MBG-Cu-TA cryogel accelerates the skin wound repair in the mice with the bacterial contamination at the injury site. In sum, GP@MBG-Cu-TA cryogel is not only endowed with dual function of hemostatic and antimicrobial capability, but also can stop bleeding of the objects with either normal or abnormal coagulation function. Thus, GP@MBG-Cu-TA cryogel provides a promising candidate dressing for managing bleeding and bacterial complications in clinic.
Collapse
Affiliation(s)
- Qixiu Hou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215500, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, 215000, China
| | - Xu He
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Suzhou, 215031, China
| | - Mengting Guo
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215500, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, 215000, China
| | - Xueqian Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215500, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, 215000, China
| | - Ziyan Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215500, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, 215000, China
| | - Xiaoyan Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215500, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, 215000, China
| | - Yong Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Suzhou, 215031, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, 215000, China.
| | - Qin Shi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Suzhou, 215031, China.
| | - Yue Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215500, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, China.
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, 215000, China.
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
24
|
Li B, Li C, Yan Z, Yang X, Xiao W, Zhang D, Liu Z, Liao X. A review of self-healing hydrogels for bone repair and regeneration: Materials, mechanisms, and applications. Int J Biol Macromol 2025; 287:138323. [PMID: 39645113 DOI: 10.1016/j.ijbiomac.2024.138323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Bone defects, which arise from various factors such as trauma, tumor resection, and infection, present a significant clinical challenge. There is an urgent need to develop new biomaterials capable of repairing a wide array of damage and defects in bone tissue. Self-healing hydrogels, a groundbreaking advancement in the field of biomaterials, displaying remarkable ability to regenerate damaged connections after partial severing, thus offering a promising solution for bone defect repair. This review first presents a comprehensive overview of the progress made in the design and preparation of these hydrogels, focusing on the self-healing mechanisms based on physical non-covalent interactions and dynamic chemical covalent bonds. Subsequently, the applications of self-healing hydrogels including natural polymers, synthetic polymers, and nano-hybrid materials, are discussed in detail, emphasizing their mechanisms in promoting bone tissue regeneration. Finally, the review addresses current challenges as well as future prospects for the use of hydrogels in bone repair and regeneration, identifying osteogenic properties, mechanical performance, and long-term biocompatibility as key areas for further improvement. In summary, this paper provides an in-depth analysis of recent advances in self-healing hydrogels for bone repair and regeneration, underscoring their immense potential for clinical application.
Collapse
Affiliation(s)
- Bo Li
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Chenchen Li
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Ziyi Yan
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Xiaoling Yang
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Wenqian Xiao
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Dawei Zhang
- Department of Orthopedics, The 960th Hospital of the PLA Joint Logistice Support Force, Jinan 250031, China.
| | - Zhongning Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China.
| | - Xiaoling Liao
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| |
Collapse
|
25
|
Chen M, Qiao Y, Yu L, Wang W, Wang W, Sun H, Xu Y, Bai J, Zhou J, Geng D. A microenvironment responsive polyetheretherketone implant with antibacterial and osteoimmunomodulatory properties facilitates osseointegration. Bioact Mater 2025; 43:273-291. [PMID: 39399839 PMCID: PMC11470486 DOI: 10.1016/j.bioactmat.2024.09.017] [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: 06/17/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Failure of intraosseous prostheses is primarily attributed to implant loosening and infections. Current primary therapeutic modalities, such as antibiotics and local debridement, not only face challenges in thoroughly eliminating obstinate adhered bacteria but also encounter difficulties in ameliorating undue inflammatory reactions and regenerating impaired peri-implant bone tissues. Polyetheretherketone (PEEK) has excellent mechanical and physicochemical characteristics and has been used extensively as a medical biomaterial. However, the limited bactericidal and osseointegrative activities of bioinert PEEK restrict its clinical application. Herein, a microenvironment responsive coating with immobilised immunomodulatory magnesium ions (Mg2+) and disinfectant cerium oxide nanoparticles (CNPs) is designed via ion coordination mediated by polydopamine (PDA) and electrospinning based on collagen structure-bionic silk fibroin (SF). By utilising the pH responsiveness of SF, CNPs exhibit potent antibacterial effects in an acidic environment (pH 5.0) caused by local bacterial infection. Due to the chelation interaction with PDA and the constraint of SF, Mg2+ is slowly released, ameliorating the local immune microenvironment and boosting osteogenesis by upregulating M2 phenotype macrophages. Bioinformatics analysis indicates that the inflammation is suppressed via the NF-κB signaling pathway. Overall, this SF-based coating maximizes the synergistic effect of CNPs and Mg2+, offering enhanced antibacterial and osteoimmunomodulatory bioactivity for successful implantation.
Collapse
Affiliation(s)
- Miao Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yusen Qiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wentao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Haifu Sun
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Jun Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
26
|
Lin C, Bai X, Zhang L. Chitosan/Sodium Tripolyphosphate/Sodium Alginate/miRNA-34c-5p Antagomir Scaffolds Promote the Functionality of Rabbit Cranial Parietal Repair. Int J Nanomedicine 2024; 19:12939-12956. [PMID: 39651352 PMCID: PMC11622683 DOI: 10.2147/ijn.s481965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 11/22/2024] [Indexed: 12/11/2024] Open
Abstract
Purpose MicroRNA-34c-5p (miR-34c-5p) plays a pivotal role in bone remodeling, yet its therapeutic potential is hindered by challenges such as instability, limited cellular internalization, and immune responses. This study was aimed at developing innovative scaffolds capable of efficiently delivering microRNAs (miRNAs), specifically miR-34c-5p. Methods Chitosan (CS)/sodium tripolyphosphate (STPP)/sodium alginate (SA) scaffolds, referred to as CTS scaffolds, were synthesized at a specific ratio and characterized using dynamic light scattering and scanning electron microscopy (SEM). Cytotoxicity assessments were conducted through cell activity staining. The loading capacity and releasing performance of miRNAs were quantified using spectrophotometry. Subsequently, the in vivo efficacy of miR-34c-5p Agomir/Antagomir in regulating bone repair was evaluated in the rabbit cranial bone defect model, with micro-CT scanning and histological analysis conducted at 4, 8, and 12 weeks. Results CTS scaffolds with a composition ratio of 1:0.2:0.1 were successfully synthesized, exhibiting a mean particle size of 360.1 nm. SEM revealed scaffolds had the porous spongy structure. Cell activity staining confirmed the excellent biocompatibility of the CTS scaffolds. Spectrophotometry demonstrated miR-34c-5p Antagomir were continually released, reaching 91.41% within 30 days. Differential new bone formation was observed between the miR-34c-5p Agomir and Antagomir groups. Micro-CT imaging and histological staining revealed varying degrees of bone regeneration, with notable improvements in the miR-34c-5p Antagomir group. Conclusion CTS scaffolds with a composition ratio of 1:0.2:0.1 demonstrate favorable biocompatibility and enable efficient loading and sustained release of miR-34c-5p Antagomir. The study suggests potential applications of miR-34c-5p Antagomir in promoting bone repair and highlights the promise of innovative scaffolds for therapeutic miRNAs administration in bone regeneration.
Collapse
Affiliation(s)
- Chen Lin
- Department of Orthodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, People’s Republic of China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| | - Xinyi Bai
- Department of Stomatology, Beijing Shunyi District Hospital, Beijing, 101300, People’s Republic of China
| | - Linkun Zhang
- Department of Orthodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, People’s Republic of China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| |
Collapse
|
27
|
Wu M, Zhang Y, Zhao Y, Chu L, Meng X, Ye L, Li X, Wang Z, Wu P. Photoactivated Hydrogel Therapeutic System with MXene-Based Nanoarchitectonics Potentiates Endogenous Bone Repair Through Reshaping the Osteo-Vascularization Network. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403003. [PMID: 39377343 DOI: 10.1002/smll.202403003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/23/2024] [Indexed: 10/09/2024]
Abstract
The repair and reconstruction of large-scale bone defects face enormous challenges because of the failure to reconstruct the osteo-vascularization network. Herein, a near-infrared (NIR) light-responsive hydrogel system is reported to achieve programmed tissue repair and regeneration through the synergetic effects of on-demand drug delivery and mild heat stimulation. The spatiotemporal hydrogel system (HG/MPa) composed of polydopamine-coated Ti3C2Tx MXene (MP) nanosheets decorated with acidic fibroblast growth factor (aFGF, a potent angiogenic drug) and hydroxypropyl chitosan/gelatin (HG) hydrogel is developed to orchestrate the reconstruction of the osteo-vascularization network and boost bone regeneration. Upon exposure to NIR light irradiation, the engineered HG/MPa hydrogel can achieve the initial complete release of aFGF to induce rapid angiogenesis and provide sufficient blood supply, maximizing its biofunction in the defect area. This integrated hydrogel system demonstrated good therapeutic efficacy in promoting cell adhesion, proliferation, migration, angiogenesis, and osteogenic differentiation through periodic NIR irradiation. In vivo, animal experiments further revealed that the spatiotemporalized hydrogel platform synergized with mild photothermal treatment significantly accelerated critical-sized bone defect healing by increasing the osteo-vascularization network density, recruiting endogenous stem cells, and facilitating the production of osteogenesis/angiogenesis-related factors. Overall, smart-responsive hydrogel could enhance the reconstruction of the osteo-vascularization network in bone regeneration.
Collapse
Affiliation(s)
- Minhao Wu
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan, 430071, China
| | - Yufeng Zhang
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yanfang Zhao
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Liuxi Chu
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaolei Meng
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Luyang Ye
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaokun Li
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhouguang Wang
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ping Wu
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
28
|
Chen X, Xu Z, Gao Y, Chen Y, Yin W, Liu Z, Cui W, Li Y, Sun J, Yang Y, Ma W, Zhang T, Tian T, Lin Y. Framework Nucleic Acid-Based Selective Cell Catcher for Endogenous Stem Cell Recruitment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406118. [PMID: 39543443 DOI: 10.1002/adma.202406118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/23/2024] [Indexed: 11/17/2024]
Abstract
Cell-surface engineering holds great promise in boosting endogenous stem cell attraction for tissue regeneration. However, challenges such as cellular internalization of ligand and the dynamic nature of cell membranes often complicate ligand-receptor interactions. The aim of this study is to harness the innovative potential of programmable tetrahedral framework nucleic acid (tFNA) to enable precise, tunable ligand-receptor interactions, thereby improving stem cell recruitment efficiency. This approach involves experimental screening and theoretical analysis using dissipative particle dynamics. The results demonstrate that altering the flexibility and topology of ligands on tFNA changes their cellular internalization and membrane binding efficiency. Furthermore, optimizing the distribution of the mesenchymal stem cell (MSC)-binding aptamer 19S (Apt19S) on the tFNA enhances the stem cell capture efficiency. Following successful in vitro MSC capture, Apt19S-modified tFNA is chemically linked to a hyaluronic acid hydrogel, forming an efficient "stem cell catcher" system. Subsequent in vivo experiments demonstrate that this system effectively promotes early stem cell recruitment and accelerates bone regeneration in different bone healing scenarios, including cranial and maxillary defects.
Collapse
Affiliation(s)
- Xingyu Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Ziang Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yang Gao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Ye Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Wumeng Yin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Weitong Cui
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yong Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Jiafei Sun
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yuting Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
29
|
Wang X, Zeng J, Gan D, Ling K, He M, Li J, Lu Y. Recent Strategies and Advances in Hydrogel-Based Delivery Platforms for Bone Regeneration. NANO-MICRO LETTERS 2024; 17:73. [PMID: 39601916 PMCID: PMC11602938 DOI: 10.1007/s40820-024-01557-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/01/2024] [Indexed: 11/29/2024]
Abstract
Bioactive molecules have shown great promise for effectively regulating various bone formation processes, rendering them attractive therapeutics for bone regeneration. However, the widespread application of bioactive molecules is limited by their low accumulation and short half-lives in vivo. Hydrogels have emerged as ideal carriers to address these challenges, offering the potential to prolong retention times at lesion sites, extend half-lives in vivo and mitigate side effects, avoid burst release, and promote adsorption under physiological conditions. This review systematically summarizes the recent advances in the development of bioactive molecule-loaded hydrogels for bone regeneration, encompassing applications in cranial defect repair, femoral defect repair, periodontal bone regeneration, and bone regeneration with underlying diseases. Additionally, this review discusses the current strategies aimed at improving the release profiles of bioactive molecules through stimuli-responsive delivery, carrier-assisted delivery, and sequential delivery. Finally, this review elucidates the existing challenges and future directions of hydrogel encapsulated bioactive molecules in the field of bone regeneration.
Collapse
Affiliation(s)
- Xiao Wang
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China
| | - Jia Zeng
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China
| | - Donglin Gan
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China
| | - Kun Ling
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China
| | - Mingfang He
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, People's Republic of China.
| | - Yongping Lu
- Scientific and Technological Innovation Center for Biomedical Materials and Clinical Research, Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, People's Republic of China.
| |
Collapse
|
30
|
Cui X, Wu L, Zhang C, Li Z. Implantable Self-Powered Systems for Electrical Stimulation Medical Devices. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2412044. [PMID: 39587936 DOI: 10.1002/advs.202412044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/27/2024] [Indexed: 11/27/2024]
Abstract
With the integration of bioelectronics and materials science, implantable self-powered systems for electrical stimulation medical devices have emerged as an innovative therapeutic approach, garnering significant attention in medical research. These devices achieve self-powering through integrated energy conversion modules, such as triboelectric nanogenerators (TENGs) and piezoelectric nanogenerators (PENGs), significantly enhancing the portability and long-term efficacy of therapeutic equipment. This review delves into the design strategies and clinical applications of implantable self-powered systems, encompassing the design and optimization of energy harvesting modules, the selection and fabrication of adaptable electrode materials, innovations in systematic design strategies, and the extensive utilization of implantable self-powered systems in biological therapies, including the treatment of neurological disorders, tissue regeneration engineering, drug delivery, and tumor therapy. Through a comprehensive analysis of the latest research progress, technical challenges, and future directions in these areas, this paper aims to provide valuable insights and inspiration for further research and clinical applications of implantable self-powered systems.
Collapse
Affiliation(s)
- Xi Cui
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Li Wu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Engineering, Chinese Academy of Sciences, Beijing, 100049, China
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Engineering, Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
31
|
Pan S, Li Y, Wang L, Guan Y, Xv K, Li Q, Feng G, Hu Y, Lan X, Qin S, Gui L, Li L. Microenvironment-optimized gastrodin-functionalized scaffolds orchestrate asymmetric division of recruited stem cells in endogenous bone regeneration. J Nanobiotechnology 2024; 22:722. [PMID: 39563380 DOI: 10.1186/s12951-024-02886-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/30/2024] [Indexed: 11/21/2024] Open
Abstract
The regeneration of osteoporotic bone defects remains challenging as the critical stem cell function is impaired by inflammatory microenvironment. Synthetic materials that intrinsically direct osteo-differentiation versus self-renewal of recruited stem cell represent a promising alternative strategy for endogenous bone formation. Therefore, a microenvironmentally optimized polyurethane (PU) /n-HA scaffold to enable sustained delivery of gastrodin is engineered to study its effect on the osteogenic fate of stem cells. It exhibited interconnected porous networks and an elevated sequential gastrodin release pattern to match immune-osteo cascade concurrent with progressive degradation of materials. In a critical-sized femur defect model of osteoporotic rat, 5% gastrodin-PU/n-HA potently promoted neo-bone regeneration by facilitating M2 macrophage polarization and CD146+ host stem cell recruitment to defective site. The implantation time-dependently increased the bone marrow mesenchymal stem cell (BMSC) population, and further culture of BMSCs showed a robust ability of proliferation, migration, and mitochondrial resurgence. Of note, some of cell pairs produced one stemness daughter cell while the other committed to osteogenic lineage in an asymmetric cell division (ACD) manner, and a much more compelling ACD response was triggered when 5% gastrodin-PU/n-HA implanted. Further investigation revealed that one-sided concentrated presentation of aPKC and β-catenin in dividing cells effectively induced asymmetric distribution, which polarized aPKC biased the response of the daughter cells to Wnt signal. The asymmetric cell division in skeletal stem cells (SSCs) was mechanically comparable to BMSCs and also governed by distinct aPKC and β-catenin biases. Concomitantly, delayed bone loss adjacent to the implant partly alleviated development of osteoporosis. In conclusion, our findings provide insight into the regulation of macrophage polarization combined with osteogenic commitment of recruited stem cells in an ACD manner, advancing scaffold design strategy for endogenous bone regeneration.
Collapse
Affiliation(s)
- Shilin Pan
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, School of Rehabilitation, Kunming Medical University, Kunming, 650500, China
| | - Yao Li
- Department of Stomatology, The First People's Hospital of Yunnan Province, Kunming, 650032, China
| | - Lu Wang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, School of Rehabilitation, Kunming Medical University, Kunming, 650500, China
| | - Yingchao Guan
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, School of Rehabilitation, Kunming Medical University, Kunming, 650500, China
| | - Kaiyang Xv
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, School of Rehabilitation, Kunming Medical University, Kunming, 650500, China
| | - Qing Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, School of Rehabilitation, Kunming Medical University, Kunming, 650500, China
| | - Guangli Feng
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| | - Yingrui Hu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, School of Rehabilitation, Kunming Medical University, Kunming, 650500, China
| | - Xiaoqian Lan
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| | - Shiyi Qin
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| | - Li Gui
- Department of Endocrinology, The Third People's Hospital of Yunnan Province, Kunming, 650011, China.
| | - Limei Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, School of Rehabilitation, Kunming Medical University, Kunming, 650500, China.
| |
Collapse
|
32
|
Wang Y, Chen Y, Zhou T, Li J, Zhang N, Liu N, Zhou P, Mao Y. A novel multifunctional nanocomposite hydrogel orchestrates the macrophage reprogramming-osteogenesis crosstalk to boost bone defect repair. J Nanobiotechnology 2024; 22:702. [PMID: 39533396 PMCID: PMC11558876 DOI: 10.1186/s12951-024-02996-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
Repairing bone defects is a complex cascade reaction process, as immune system regulation, vascular growth, and osteogenic differentiation are essential. Thus, developing a tissue-engineered biomaterial that caters to the complex healing process of bone regeneration remains a major clinical challenge. In the study, Ca2+-TA-rGO (CTAG)/GelMA hydrogels were synthesized by binding Ca2+ using metal chelation to graphene oxide (GO) nanosheets reduced by tannic acid (TA-rGO) and doping them into gelatin methacrylate (GelMA) hydrogels. TA and rGO exhibited biocompatibility and immunomodulatory properties in this composite, while Ca2+ promoted bone formation and angiogenesis. This novel nanocomposite hydrogel demonstrated good mechanical properties, degradability, and conductivity, and it could achieve slow Ca2+ release during bone regeneration. Both in vitro and in vivo experiments revealed that CTAG/GelMA hydrogel modulated macrophage reprogramming and induced a shift from macrophages to healing-promoting M2 macrophages during the inflammatory phase, promoted vascular neovascularization, and facilitated osteoblast differentiation during bone formation. Moreover, CTAG/GelMA hydrogel could downregulate the NF-κB signaling pathway, offering new insights into regulating macrophage reprogramming-osteogenic crosstalk. Conclusively, this novel multifunctional nanocomposite hydrogel provides a multistage treatment for bone and orchestrates macrophage reprogramming-osteogenic crosstalk to boost bone repair.
Collapse
Affiliation(s)
- Ying Wang
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Yedan Chen
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Tao Zhou
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, 233030, China
| | - Jingze Li
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, 233030, China
| | - Na Zhang
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Na Liu
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Pinghui Zhou
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China.
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, 233030, China.
| | - Yingji Mao
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China.
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China.
- Anhui Nerve Regeneration Technology and Medical New Materials Engineering Research Center, Bengbu Medical University, Bengbu, 233030, China.
| |
Collapse
|
33
|
Bai M, Wang T, Xing Z, Huang H, Wu X, Adeli M, Wang M, Han X, Ye L, Cheng C. Electron-donable heterojunctions with synergetic Ru-Cu pair sites for biocatalytic microenvironment modulations in inflammatory mandible defects. Nat Commun 2024; 15:9592. [PMID: 39505847 PMCID: PMC11541594 DOI: 10.1038/s41467-024-53824-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
The clinical treatments of maxillofacial bone defects pose significant challenges due to complex microenvironments, including severe inflammation, high levels of reactive oxygen species (ROS), and potential bacterial infection. Herein, we propose the de novo design of an efficient, versatile, and precise electron-donable heterojunction with synergetic Ru-Cu pair sites (Ru-Cu/EDHJ) for superior biocatalytic regeneration of inflammatory mandible defects and pH-controlled antibacterial therapies. Our studies demonstrate that the unique structure of Ru-Cu/EDHJ enhances the electron density of Ru atoms and optimizes the binding strength of oxygen species, thus improving enzyme-like catalytic performance. Strikingly, this biocompatible Ru-Cu/EDHJ can efficiently switch between ROS scavenging in neutral media and ROS generation in acidic media, thus simultaneously exhibiting superior repair functions and bioadaptive antibacterial properties in treating mandible defects in male mice. We believe synthesizing such biocatalytic heterojunctions with exceptional enzyme-like capabilities will offer a promising pathway for engineering ROS biocatalytic materials to treat trauma, tumors, or infection-caused maxillofacial bone defects.
Collapse
Grants
- 52161145402, 52173133, 52373148 National Natural Science Foundation of China (National Science Foundation of China)
- 82470962, 82001020 National Natural Science Foundation of China (National Science Foundation of China)
- U21A20368 National Natural Science Foundation of China (National Science Foundation of China)
- sklpme2021-4-02 State Key Laboratory of Polymer Materials Engineering
- National Key R&D Program of China (2021YFB3800700),Sichuan Science and Technology Program (2023YFH0008),the 1·3·5 Project for Disciplines of Excellence, West China Hospital, Sichuan University (ZYJC21047).
- Sichuan Science and Technology Program (2024NSFSC0672, 2021YFG0238),China Postdoctoral Science Foundation (2019M663525), Research Funding from West China School/Hospital of Stomatology Sichuan University (RCDWJS2023-16), and Research and Develop Program, West China Hospital of Stomatology Sichuan University (RD-02-202206).
- National Key R&D Program of China (2023YFC3605600), Sichuan Science and Technology Program (2023YFS0019), Med-X Innovation Programme of Med-X Center for Materials, Sichuan University (MCMGD202301)
Collapse
Affiliation(s)
- Mingru Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ting Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Zhenyu Xing
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Haoju Huang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Xizheng Wu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Mohsen Adeli
- Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Mao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Chong Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China.
| |
Collapse
|
34
|
Zhu Y, Liu H, Wu P, Chen Y, Deng Z, Cai L, Wu M. Multifunctional injectable hydrogel system as a mild photothermal-assisted therapeutic platform for programmed regulation of inflammation and osteo-microenvironment for enhanced healing of diabetic bone defects in situ. Theranostics 2024; 14:7140-7198. [PMID: 39629118 PMCID: PMC11610133 DOI: 10.7150/thno.102779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/19/2024] [Indexed: 12/06/2024] Open
Abstract
Background: Factor-free biomaterial scaffolds play an increasingly important role in promoting in situ bone reconstruction and regeneration. However, the complicated and variable pathophysiological microenvironments of the injury sites under diabetic conditions, including the vicious cycle of oxidative stress and inflammatory response, impaired osteo/angiogenesis function and hyperactive osteoclastogenesis, as well as increased susceptibility to bacterial infection, may largely weaken the therapeutic potential of implanted scaffolds, leading to uncontrolled and poor outcomes of bone defect healing. Methods and Results: To tackle the aforementioned challenges, a mild photothermal-assisted multifunctional therapeutic platform (denoted as GAD/MC) that integrates copper-containing two-dimensional Ti3C2Tx MXene nanosheets, gelatin methacrylate, and alginate-graft-dopamine was proposed to achieve efficient and synergistic therapy for diabetic bone defects. Thereinto, copper-decorated MXene (MC) nanosheets were employed as both functional crosslinkers and nanofillers to participate in the construction of an interpenetrating polymer network structure through multiple covalent and noncovalent bonds, which conferred the hydrogel with advantageous traits like enhanced mechanical properties, injectability and moldability, strong bone tissue adhesion and self-healing ability, as well as excellent anti-swelling and near-infrared (NIR) photothermal conversion capabilities. On account of the NIR/pH dual-responsive properties, the resulting hydrogel system was capable of achieving the controlled and stimuli-responsive release of bioactive Cu2+, allowing on-demand delivery at the site of injury. Moreover, with the assistance of mild photothermal effects, this integrated hydrogel system demonstrated remarkable antibacterial and antioxidant properties. It effectively scavenged excessive reactive oxygen species (ROS), inhibited inflammatory responses, and promoted macrophage polarization towards the pro-healing M2 phenotype. Such characteristics were beneficial for recreating an optimized microenvironment that supported the adhesion, proliferation, migration, and differentiation of osteoblasts and endothelial cells, while concurrently inhibiting osteoclast function. In a critical-sized cranial defect model using diabetic rats, the injectable GAD/MC hydrogel system combined with on-demand mild hyperthermia further synergistically accelerated new bone formation and bone healing processes by eliminating intracellular ROS, ameliorating inflammation, orchestrating M2 macrophage polarization, promoting osteo/angiogenesis, and suppressing osteoclastogenesis. Conclusions: Overall, the constructed multifunctional injectable hydrogel system has emerged as a promising therapeutic candidate for addressing complex bone-related challenges by remodeling the disordered immune microenvironment and expediting the bone healing process.
Collapse
Affiliation(s)
- Yufan Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan 430071 Hubei, China
| | - Huifan Liu
- Department of Anesthesiology, Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ping Wu
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yun Chen
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medicine Sciences), Wuhan University, Wuhan 430071, China
| | - Zhouming Deng
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan 430071 Hubei, China
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan 430071 Hubei, China
| | - Minhao Wu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan 430071 Hubei, China
| |
Collapse
|
35
|
Tang Y, Liu J, Feng S, Long H, Lai W, Xiang L. Exploration of bone metabolism status in the distal femur of mice at different growth stages. Biochem Biophys Res Commun 2024; 729:150351. [PMID: 38996655 DOI: 10.1016/j.bbrc.2024.150351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024]
Abstract
The mouse femur, particularly the distal femur, is commonly utilized in orthopedic research. Despite its significance, little is known about the key events involved in the postnatal development of the distal femur. Therefore, investigating the development process of the mouse distal femur is of great importance. In this study, distal femurs of CD-1 mice aged 1, 2, 4, 6, and 8 weeks were examined. We found that the width and height of the distal femur continued to increase till the 4th week, followed with stabilization. Notably, the width to height ratio remained relatively consistent with age. Micro computed tomography analysis demonstrated gradual increases in bone volume/tissue volume, trabecular number, and trabecular thickness from 1 to 6 weeks, alongside a gradual decrease in trabecular separation. Histological analysis further indicated the appearance of the secondary ossification center at approximately 2 weeks, with ossification mostly completed by 4 weeks, leading to the formation of a prototype epiphyseal plate. Subsequently, the epiphyseal plate gradually narrowed at 6 and 8 weeks. Moreover, the thickness and maturity of the bone cortex surrounding the epiphyseal plate increased over time, reaching peak cortical bone density at 8 weeks. In conclusion, to enhance model stability and operational ease, we recommend constructing conventional mouse models of the distal femur between 4 and 8 weeks old.
Collapse
Affiliation(s)
- Yufei Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, China
| | - Jiayi Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, China
| | - Shuqi Feng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, China
| | - Hu Long
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, China
| | - Wenli Lai
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, China.
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, China.
| |
Collapse
|
36
|
Zheng Y, Tan L, Chen H, He S, Li M, Luo Z, Cai K, Hu Y. Hierarchical Integration of Curcumin-Loaded CaCO 3 Nanoparticles and Black Phosphorus Nanosheets in Core/Shell Nanofiber for Cranial Defect Repair. Adv Healthc Mater 2024:e2401786. [PMID: 39375960 DOI: 10.1002/adhm.202401786] [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: 05/16/2024] [Revised: 09/15/2024] [Indexed: 10/09/2024]
Abstract
Reconstruction and healing of large craniofacial bone defects are major clinical challenges due to high risk of chronic inflammation and reduced cell mineralization levels. Herein, a core-shell nanofiber-based implant with significant pro-osteogenesis capability for treating skull defects is reported, which is hierarchically integrated with curcumin-loaded calcium carbonate nanoparticles (CaCO3@Cur NPs) in the outer layers and black phosphorus nanosheets (BPNSs) in the core compartments. The radical alignment of the integrated nanocomponents allows the sequential in situ release of the therapeutic agents in a controlled manner after implantation. Curcumin can repolarize M1 macrophages into M2 phenotypes for anti-inflammation purposes. Meanwhile, the released calcium and phosphate ions can promote the biomineralization of hydroxyapatite at the defect site and facilitate bone regeneration. Evaluations on cranial defect-bearing rat models demonstrated that the electrospun fibers in the present study substantially promoted restoration of the damaged skulls and inhibited inflammation in the wound bed. This strategy provides a new idea for the treatment of skull defects in the clinic.
Collapse
Affiliation(s)
- Yan Zheng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Lu Tan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Hang Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Shuohan He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
37
|
Yan X, An N, Zhang Z, Qiu Q, Yang D, Wei P, Zhang X, Qiu L, Guo J. Graphene Oxide Quantum Dots-Preactivated Dental Pulp Stem Cells/GelMA Facilitates Mitophagy-Regulated Bone Regeneration. Int J Nanomedicine 2024; 19:10107-10128. [PMID: 39381026 PMCID: PMC11460356 DOI: 10.2147/ijn.s480979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024] Open
Abstract
Background In bone tissue engineering (BTE), cell-laden scaffolds offer a promising strategy for repairing bone defects, particularly when host cell regeneration is insufficient due to age or disease. Exogenous stem cell-based BTE requires bioactive factors to activate these cells. Graphene oxide quantum dots (GOQDs), zero-dimensional derivatives of graphene oxide, have emerged as potential osteogenic nanomedicines. However, constructing biological scaffolds with GOQDs and elucidating their biological mechanisms remain critical challenges. Methods We utilized GOQDs with a particle size of 10 nm, characterized by a surface rich in C-O-H and C-O-C functional groups. We developed a gelatin methacryloyl (GelMA) hydrogel incorporated with GOQDs-treated dental pulp stem cells (DPSCs). These constructs were transplanted into rat calvarial bone defects to estimate the effectiveness of GOQDs-induced DPSCs in repairing bone defects while also investigating the molecular mechanism underlying GOQDs-induced osteogenesis in DPSCs. Results GOQDs at 5 μg/mL significantly enhanced the osteogenic differentiation of DPSCs without toxicity. The GOQDs-induced DPSCs showed active osteogenic potential in three-dimensional cell culture system. In vivo, transplantation of GOQDs-preactivated DPSCs/GelMA composite effectively facilitated calvarial bone regeneration. Mechanistically, GOQDs stimulated mitophagy flux through the phosphatase-and-tensin homolog-induced putative kinase 1 (PINK1)/Parkin E3 ubiquitin ligase (PRKN) pathway. Notably, inhibiting mitophagy with cyclosporin A prevented the osteogenic activity of GOQDs. Conclusion This research presents a well-designed bionic GOQDs/DPSCs/GelMA composite scaffold and demonstrated its ability to promote bone regeneration by enhancing mitophagy. These findings highlight the significant potential of this composite for application in BTE and underscore the crucial role of mitophagy in promoting the osteogenic differentiation of GOQDs-induced stem cells.
Collapse
Affiliation(s)
- Xiaoyuan Yan
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Na An
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Zeying Zhang
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Qiujing Qiu
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Di Yang
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Penggong Wei
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Xiyue Zhang
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Lihong Qiu
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Jiajie Guo
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| |
Collapse
|
38
|
Wang P, Wu J, Xiao X, Fan Y, Han X, Sun Y. Engineering Injectable Coassembled Hydrogel by Photothermal Driven Chitosan-Stabilized MoS 2 Nanosheets for Infected Wound Healing. ACS NANO 2024; 18:26961-26974. [PMID: 39305262 DOI: 10.1021/acsnano.4c08883] [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: 10/02/2024]
Abstract
The application of enzyme-like molybdenum disulfide (MoS2) in tissue repair was confronted with stable dispersion, solubilization, and biotoxicity. Here, the injectable self-healing hydrogel was successfully designed using a step-by-step coassembly of chitosan and MoS2. Polyphenolic chitosan as a "structural stabilizer" of MoS2 nanosheets reconstructed well-dispersed MoS2@CSH nanosheets, which improved the biocompatibility of traditional MoS2, and strengthened its photothermal conversion and enzyme-like activities, guaranteeing highly efficient radical scavenging and antimicrobial properties. Furthermore, the polyphenol chitosan was employed again as a "molecular cross-linking agent" to form the injectable NIR-responsive MoS2@CSH hydrogel by accelerating hydrogen-bond interaction among chitosan and the multicross-linking reaction among polyphenols. The rapid self-healing ability was conducive to wound closure and dynamic adaptability. An experimental study on infected wound healing demonstrated that MoS2@CSH hydrogel could substantially eradicate bacteria and accelerate the angiogenesis of infected wounds. The photothermal-driven coassembly of MoS2 and polycation provided an alternative strategy for infected wound healing.
Collapse
Affiliation(s)
- Peilei Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
| | - Jingwen Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiaolin Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
| |
Collapse
|
39
|
Gao W, Miao X, Xu T. Wilms tumor 1-associated protein mediated m6A modification promotes osteogenic differentiation of stem cells from human exfoliated deciduous teeth. J Dent Sci 2024; 19:2305-2314. [PMID: 39347097 PMCID: PMC11437296 DOI: 10.1016/j.jds.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 02/20/2024] [Indexed: 10/01/2024] Open
Affiliation(s)
- Weiheng Gao
- Department of Emergency, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center for Child Health, Hangzhou, China
| | - Xixi Miao
- National Clinical Research Center for Child Health, Hangzhou, China
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Xu
- Department of Stomatology, Nanjing Geriatric Hospital, Nanjing, China
| |
Collapse
|
40
|
Yang Y, Wang W, Zeng Q, Wang N, Li W, Chen B, Guan Q, Li C, Li W. Fabricating oxygen self-supplying 3D printed bioactive hydrogel scaffold for augmented vascularized bone regeneration. Bioact Mater 2024; 40:227-243. [PMID: 38973993 PMCID: PMC11226730 DOI: 10.1016/j.bioactmat.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/26/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
Limited cells and factors, inadequate mechanical properties, and necrosis of defects center have hindered the wide clinical application of bone-tissue engineering scaffolds. Herein, we construct a self-oxygenated 3D printed bioactive hydrogel scaffold by integrating oxygen-generating nanoparticles and hybrid double network hydrogel structure. The hydrogel scaffold possesses the characteristics of extracellular matrix; Meanwhile, the fabricated hybrid double network structure by polyacrylamide and CaCl2-crosslinked sodium carboxymethylcellulose endows the hydrogel favorable compressive strength and 3D printability. Furthermore, the O2 generated by CaO2 nanoparticles encapsulated in ZIF-8 releases steadily and sustainably because of the well-developed microporous structure of ZIF-8, which can significantly promote cell viability and proliferation in vitro, as well as angiogenesis and osteogenic differentiation with the assistance of Zn2+. More significantly, the synergy of O2 and 3D printed pore structure can prevent necrosis of defects center and facilitate cell infiltration by providing cells the nutrients and space they need, which can further induce vascular network ingrowth and accelerate bone regeneration in all areas of the defect in vivo. Overall, this work provides a new avenue for preparing cell/factor-free bone-tissue engineered scaffolds that possess great potential for tissue regeneration and clinical alternative.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Wanmeng Wang
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Qianrui Zeng
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Ning Wang
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Wenbo Li
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Bo Chen
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Qingxin Guan
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Changyi Li
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Wei Li
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| |
Collapse
|
41
|
Kong L, Zhao Y, Xiong Y, Chen J, Wang S, Yan Z, Shi H, Liu Z, Wang X. Multiscale engineered artificial compact bone via bidirectional freeze-driven lamellated organization of mineralized collagen microfibrils. Bioact Mater 2024; 40:168-181. [PMID: 38910968 PMCID: PMC11192986 DOI: 10.1016/j.bioactmat.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/12/2024] [Accepted: 02/06/2024] [Indexed: 06/25/2024] Open
Abstract
Bone, renowned for its elegant hierarchical structure and unique mechanical properties, serves as a constant source of inspiration for the development of synthetic materials. However, achieving accurate replication of bone features in artificial materials with remarkable structural and mechanical similarity remains a significant challenge. In this study, we employed a cascade of continuous fabrication processes, including biomimetic mineralization of collagen, bidirectional freeze-casting, and pressure-driven fusion, to successfully fabricate a macroscopic bulk material known as artificial compact bone (ACB). The ACB material closely replicates the composition, hierarchical structures, and mechanical properties of natural bone. It demonstrates a lamellated alignment of mineralized collagen (MC) microfibrils, similar to those found in natural bone. Moreover, the ACB exhibits a similar high mineral content (70.9 %) and density (2.2 g/cm3) as natural cortical bone, leading to exceptional mechanical properties such as high stiffness, hardness, and flexural strength that are comparable to those of natural bone. Importantly, the ACB also demonstrates excellent mechanical properties in wet, outstanding biocompatibility, and osteogenic properties in vivo, rendering it suitable for a broad spectrum of biomedical applications, including orthopedic, stomatological, and craniofacial surgeries.
Collapse
Affiliation(s)
- Lingwenyao Kong
- 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, China
| | - Yonggang Zhao
- State Key Laboratory for Performance and Structure Safety of Petroleum Tubular Goods and Equipment Materials, Tubular Goods Research Institute of CNPC, Xi'an, 710077, China
| | - Yang Xiong
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junlin Chen
- 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, China
| | - Shuo 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, China
| | - Ziming Yan
- School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
| | - Huibin Shi
- School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
| | - Zhanli Liu
- School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
| | - Xiumei 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, China
| |
Collapse
|
42
|
Park SY, Lee JK, Lee SH, Kim DS, Jung JW, Kim JH, Baek SW, You S, Hwang DY, Han DK. Multifunctional vitamin D-incorporated PLGA scaffold with BMP/VEGF-overexpressed tonsil-derived MSC via CRISPR/Cas9 for bone tissue regeneration. Mater Today Bio 2024; 28:101254. [PMID: 39328787 PMCID: PMC11426062 DOI: 10.1016/j.mtbio.2024.101254] [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: 07/24/2024] [Revised: 09/08/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Guiding endogenous regeneration of bone defects using biomaterials and regenerative medicine is considered an optimal strategy. One of the effective therapeutic approaches involves using transgene-expressed stem cells to treat tissue destruction and replace damaged parts. Among the various gene editing techniques for cells, clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) is considered as a promising method owing to the increasing therapeutic potential of cells by targeting specific sites. Herein, a vitamin D-incorporated poly(lactic-co-glycolic acid) (PLGA) scaffold with bone morphogenetic protein 2 (BMP2)/vascular endothelial growth factor (VEGF)-overexpressed tonsil-derived MSCs (ToMSCs) via CRISPR/Cas9 was introduced for bone tissue regeneration. The optimized seeding ratio of engineered ToMSCs on the scaffold demonstrated favorable immunomodulatory function, angiogenesis, and osteogenic activity in vitro. The multifunctional scaffold could potentially support stem cell in vivo and induce the transition from M1 to M2 macrophage with magnesium hydroxide and vitamin D. This study highlights the improved synergistic effect of a vitamin D-incorporated PLGA scaffold and a gene-edited ToMSCs for bone tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- So-Yeon Park
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Sang-Hyeok Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Ji-Won Jung
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Seungkwon You
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Dong-Youn Hwang
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| |
Collapse
|
43
|
Yang S, Wu H, Peng C, He J, Pu Z, Lin Z, Wang J, Hu Y, Su Q, Zhou B, Yong X, Lan H, Hu N, Hu X. From the microspheres to scaffolds: advances in polymer microsphere scaffolds for bone regeneration applications. BIOMATERIALS TRANSLATIONAL 2024; 5:274-299. [PMID: 39734699 PMCID: PMC11681185 DOI: 10.12336/biomatertransl.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 12/31/2024]
Abstract
The treatment and repair of bone tissue damage and loss due to infection, tumours, and trauma are major challenges in clinical practice. Artificial bone scaffolds offer a safer, simpler, and more feasible alternative to bone transplantation, serving to fill bone defects and promote bone tissue regeneration. Ideally, these scaffolds should possess osteoconductive, osteoinductive, and osseointegrative properties. However, the current first-generation implants, represented by titanium alloys, have shown poor bone-implant integration performance and cannot meet the requirements for bone tissue repair. This has led to increased research on second and third generation artificial bone scaffolds, which focus on loading bioactive molecules and cells. Polymer microspheres, known for their high specific surface areas at the micro- and nanoscale, exhibit excellent cell and drug delivery behaviours. Additionally, with their unique rigid structure, microsphere scaffolds can be constructed using methods such as thermal sintering, injection, and microsphere encapsulation. These scaffolds not only ensure the excellent cell drug loading performance of microspheres but also exhibit spatial modulation behaviour, aiding in bone repair within a three-dimensional network structure. This article provides a summary and discussion of the use of polymer microsphere scaffolds for bone repair, focusing on the mechanisms of bone tissue repair and the current status of clinical bone grafts, aimed at advancing research in bone repair.
Collapse
Affiliation(s)
- Shuhao Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Haoming Wu
- School of Preclinical Medicine of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Chao Peng
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Jian He
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Zhengguang Pu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Zhidong Lin
- The Second Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Jun Wang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yingkun Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Qiao Su
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Bingnan Zhou
- School of Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Yong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan Province, China
| | - Hai Lan
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
44
|
Song T, Zhao F, Yan L, Liu P, Yang J, Ruan C, Li D, Xiao Y, Zhang X. Structure driven bio-responsive ability of injectable nanocomposite hydrogels for efficient bone regeneration. Biomaterials 2024; 309:122601. [PMID: 38713973 DOI: 10.1016/j.biomaterials.2024.122601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/27/2024] [Accepted: 05/02/2024] [Indexed: 05/09/2024]
Abstract
Injectable hydrogels are promising for treatment of bone defects in clinic owing to their minimally invasive procedure. Currently, there is limited emphasis on how to utilize injectable hydrogels to mobilize body's regenerative potential for enhancing bone regeneration. Herein, an injectable bone-mimicking hydrogel (BMH) scaffold assembled from nanocomposite microgel building blocks was developed, in which a highly interconnected microporous structure and an inorganic/organic (methacrylated hydroxyapatite and methacrylated gelatin) interweaved nano structure were well-designed. Compared with hydrogels lacking micro-nano structures or only showing microporous structure, the BMH scaffold enhanced the ingrowth of vessels and promoted the formation of dense cellular networks (including stem cells and M2 macrophages), across the entire scaffold at early stage after subcutaneous implantation. Moreover, the BMH scaffold could not only directly trigger osteogenic differentiation of the infiltrated stem cells, but also provided an instructive osteo-immune microenvironment by inducing macrophages into M2 phenotype. Mechanistically, our results reveal that the nano-rough structure of the BMH plays an essential role in inducing macrophage M2 polarization through activating mechanotransduction related RhoA/ROCK2 pathway. Overall, this work offers an injectable hydrogel with micro-nano structure driven bio-responsive abilities, highlighting harnessing body's inherent regenerative potential to realize bone regeneration.
Collapse
Affiliation(s)
- Tao Song
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Fengxin Zhao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Ling Yan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Puxin Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Jirong Yang
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedical and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Changshun Ruan
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedical and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dongxiao Li
- Sichuan Academy of Chinese Medicine Science, Chengdu, 610042, China
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
45
|
Liu Y, Li L, He M, Xu Y, Wu Z, Xu X, Luo K, Lv H. Self-assembled peptide hydrogel loaded with functional peptide Dentonin accelerates vascularized bone tissue regeneration in critical-size bone defects. Regen Biomater 2024; 11:rbae106. [PMID: 39263324 PMCID: PMC11387769 DOI: 10.1093/rb/rbae106] [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: 05/26/2024] [Revised: 07/18/2024] [Accepted: 08/18/2024] [Indexed: 09/13/2024] Open
Abstract
Regeneration of oral craniofacial bone defects is a complex process, and reconstruction of large bone defects without the use of exogenous cells or bioactive substances remains a major challenge. Hydrogels are highly hydrophilic polymer networks with the potential to promote bone tissue regeneration. In this study, functional peptide Dentonin was loaded onto self-assembled peptide hydrogels (RAD) to constitute functionally self-assembling peptide RAD/Dentonin hydrogel scaffolds with a view that RAD/Dentonin hydrogel could facilitate vascularized bone regeneration in critical-size calvarial defects. The functionalized peptide RAD/Dentonin forms highly ordered β-sheet supramolecular structures via non-covalent interactions like hydrogen bonding, ultimately assembling into nano-fiber network. RAD/Dentonin hydrogels exhibited desirable porosity and swelling properties, and appropriate biodegradability. RAD/Dentonin hydrogel supported the adhesion, proliferation and three-dimensional migration of bone marrow mesenchymal stem cells (BMSCs) and has the potential to induce differentiation of BMSCs towards osteogenesis through activation of the Wnt/β-catenin pathway. Moreover, RAD/Dentonin hydrogel modulated paracrine secretion of BMSCs and increased the migration, tube formation and angiogenic gene expression of human umbilical vein endothelial cells (HUVECs), which boosted the angiogenic capacity of HUVECs. In vivo, RAD/Dentonin hydrogel significantly strengthened vascularized bone formation in rat calvarial defect. Taken together, these results indicated that the functionalized self-assembling peptide RAD/Dentonin hydrogel effectively enhance osteogenic differentiation of BMSCs, indirectly induce angiogenic effects in HUVECs, and facilitate vascularized bone regeneration in vivo. Thus, it is a promising bioactive material for oral and maxillofacial regeneration.
Collapse
Affiliation(s)
- Yijuan Liu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Li Li
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Mengjiao He
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Yanmei Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Zekai Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Hongbing Lv
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| |
Collapse
|
46
|
Lin J, Jia S, Cao F, Huang J, Chen J, Wang J, Liu P, Zeng H, Zhang X, Cui W. Research Progress on Injectable Microspheres as New Strategies for the Treatment of Osteoarthritis Through Promotion of Cartilage Repair. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202400585] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Indexed: 07/07/2024]
Abstract
AbstractOsteoarthritis (OA) is a degenerative disease caused by a variety of factors with joint pain as the main symptom, including fibrosis, chapping, ulcers, and loss of cartilage. Traditional treatment can only delay the progression of OA, and classical delivery system have many side effects. In recent years, microspheres have shown great application prospects in the field of OA treatment. Microspheres can support cells, reproduce the natural tissue microenvironment in vitro and in vivo, and are an efficient delivery system for the release of drugs or biological agents, which can promote cell proliferation, migration, and differentiation. Thus, they have been widely used in cartilage repair and regeneration. In this review, preparation processes, basic materials, and functional characteristics of various microspheres commonly used in OA treatment are systematically reviewed. Then it is introduced surface modification strategies that can improve the biological properties of microspheres and discussed a series of applications of microsphere functionalized scaffolds in OA treatment. Finally, based on bibliometrics research, the research development, future potential, and possible research hotspots of microspheres in the field of OA therapy is systematically and dynamically evaluated. The comprehensive and systematic review will bring new understanding to the field of microsphere treatment of OA.
Collapse
Affiliation(s)
- Jianjing Lin
- Department of Sports Medicine and Rehabilitation Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
| | - Shicheng Jia
- Department of Sports Medicine and Rehabilitation Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
- Shantou University Medical College Shantou Guangdong 515041 P. R. China
| | - Fuyang Cao
- Department of Orthopedics Second Hospital of Shanxi Medical University Taiyuan Shanxi 030001 P. R. China
| | - Jingtao Huang
- Shantou University Medical College Shantou Guangdong 515041 P. R. China
| | - Jiayou Chen
- Department of Sports Medicine and Rehabilitation Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
- Shantou University Medical College Shantou Guangdong 515041 P. R. China
| | - Juan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200025 P. R. China
| | - Peng Liu
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
| | - Hui Zeng
- Shenzhen Second People's Hospital (First Affiliated Hospital of Shenzhen University) Shenzhen Guangdong 518035 China
| | - Xintao Zhang
- Department of Sports Medicine and Rehabilitation Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200025 P. R. China
| |
Collapse
|
47
|
Li X, Wang Y, Zhao C, Tong L, Wang P, Liang J, Jiang Q, Fan Y, Zhang X, Sun Y. A facile Immunoregulatory Constructional Design by Proanthocyanidin Optimizing Directional Chitosan Microchannel. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310689. [PMID: 38421135 DOI: 10.1002/smll.202310689] [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: 11/21/2023] [Revised: 02/05/2024] [Indexed: 03/02/2024]
Abstract
Improving the interconnected structure and bioregulatory function of natural chitosan is beneficial for optimizing its performance in bone regeneration. Here, a facile immunoregulatory constructional design is proposed for developing instructive chitosan by directional freezing and alkaline salting out. The molecular dynamics simulation confirmed the assembly kinetics and structural features of various polyphenols and chitosan molecules. Along with the in vitro anti-inflammatory, antioxidative, promoting bone mesenchymal stem cell (BMSC) adhesion and proliferation performance, proanthocyanidin optimizing chitosan (ChiO) scaffold presented an optimal immunoregulatory structure with the directional microchannel. Transcriptome analysis in vitro further revealed the cytoskeleton- and immune-regulation effect of ChiO are the key mechanism of action on BMSC. The rabbit cranial defect model (Φ = 10 mm) after 12 weeks of implantation confirmed the significantly enhanced bone reconstitution. This facile immunoregulatory directional microchannel design provides effective guidance for developing inducible chitosan scaffolds.
Collapse
Affiliation(s)
- Xing Li
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Yuxiang Wang
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Chengkun Zhao
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Lei Tong
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Peilei Wang
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Jie Liang
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29# Wangjiang Road, Chengdu, 610064, P. R. China
| | - Qing Jiang
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Yujiang Fan
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Xingdong Zhang
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Yong Sun
- CAE/NAE, National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| |
Collapse
|
48
|
Jiang W, Zhan Y, Zhang Y, Sun D, Zhang G, Wang Z, Chen L, Sun J. Synergistic large segmental bone repair by 3D printed bionic scaffolds and engineered ADSC nanovesicles: Towards an optimized regenerative microenvironment. Biomaterials 2024; 308:122566. [PMID: 38603824 DOI: 10.1016/j.biomaterials.2024.122566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/08/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Achieving sufficient bone regeneration in large segmental defects is challenging, with the structure of bone repair scaffolds and their loaded bioactive substances crucial for modulating the local osteogenic microenvironment. This study utilized digital laser processing (DLP)-based 3D printing technology to successfully fabricate high-precision methacryloylated polycaprolactone (PCLMA) bionic bone scaffold structures. Adipose-derived stem cell-engineered nanovesicles (ADSC-ENs) were uniformly and stably modified onto the bionic scaffold surface using a perfusion device, constructing a conducive microenvironment for tissue regeneration and long bone defect repair through the scaffold's structural design and the vesicles' biological functions. Scanning electron microscopy (SEM) examination of the scaffold surface confirmed the efficient loading of ADSC-ENs. The material group loaded with vesicles (PCLMA-BAS-ENs) demonstrated good cell compatibility and osteogenic potential when analyzed for the adhesion and osteogenesis of primary rabbit bone marrow mesenchymal stem cells (BMSCs) on the material surface. Tested in a 15 mm critical rabbit radial defect model, the PCLMA-BAS-ENs scaffold facilitated near-complete bone defect repair after 12 weeks. Immunofluorescence and proteomic results indicated that the PCLMA-BAS-ENs scaffold significantly improved the osteogenic microenvironment at the defect site in vivo, promoted angiogenesis, and enhanced the polarization of macrophages towards M2 phenotype, and facilitated the recruitment of BMSCs. Thus, the PCLMA-BAS-ENs scaffold was proven to significantly promote the repair of large segmental bone defects. Overall, this strategy of combining engineered vesicles with highly biomimetic scaffolds to promote large-segment bone tissue regeneration holds great potential in orthopedic and other regenerative medicine applications.
Collapse
Affiliation(s)
- Wenbin Jiang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Yichen Zhan
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Yifan Zhang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Di Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Guo Zhang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Zhenxing Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Lifeng Chen
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China.
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China.
| |
Collapse
|
49
|
Huang Y, Hu R, Liu Z, Geng Y, Li F, Song Y, Ma W, Dong H, Xu L, Zhang M, Song K. Bushen Huoxue recipe ameliorates ovarian function via promoting BMSCs proliferation and homing to ovaries in POI mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155630. [PMID: 38678952 DOI: 10.1016/j.phymed.2024.155630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/30/2024] [Accepted: 04/11/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is a tricky puzzle in the field of female reproductive medicine. Bushen Huoxue recipe (BHR), a traditional Chinese medicine compound based on the combination of kidney-tonifying and blood-activating functions, has shown excellent efficacy in improving female irregular menstruation, POI, and infertility. However, the potential mechanism of BHR in POI treatment has not yet been elucidated. Bone marrow mesenchymal stem cells (BMSCs), a type of pluripotent stem cells, have received increasing attention for their significant role in improving ovarian function and restoring fertility in women with POI. PURPOSE This study aimed to evaluate the therapeutic effect of BHR in POI mice and explore its potential mechanism. METHODS A POI mouse model was established with a single intraperitoneal injection of 120 mg/kg cyclophosphamide (CTX). Distilled water, BHR, or dehydroepiandrosterone was administered via gavage for 28 consecutive days. The effect of BHR on ovarian function in POI mice was evaluated by assessing the estrous cycle, ovarian morphology, follicular development, hormone levels, and angiogenesis. The proportion of BMSCs in bone marrow, peripheral blood, and ovary was analyzed via flow cytometry, and the level of molecules mediating migration and homing in ovary was measured. Cell viability assays, scratch healing assays and transwell migration assays were performed to explore the effect of BHR on BMSCs proliferation and migration in vitro, and its potential mechanism was explored. RESULTS BHR significantly ameliorated estrous cycle disorders, hormone disorders, ovarian morphology, ovarian microvascular formation, and ovarian reserve in POI mice. Meanwhile, the number of BMSCs number in the bone marrow, peripheral blood, and ovary was apparently increased. Of note, BHR increased the level of hepatocyte growth factor (HGF)/cellular mesenchymal epithelial transition factor (cMET) and stromal cell-derived factor-1(SDF-1)/CXC chemokine receptor 4 (CXCR4) in the ovaries of POI mice. Moreover, BHR treatment promoted BMSCs proliferation and migration in vitro, with a significant increase in the level of proliferating cell nuclear antigen, cMET, and CXCR4. CONCLUSIONS BHR effectively restored ovarian reserve, ovarian function, and ovarian angiogenesis in CTX-induced POI mice. In addition, BHR promoted BMSCs proliferation, migration, and homing to the ovary, which was mediated by the SDF-1/CXCR4 and HGF/cMET signaling axis. Finally, the amelioration of ovarian reserve and ovarian function in CTX-induced POI mice by BHR may be related to its promotion of endogenous BMSCs proliferation and homing.
Collapse
Affiliation(s)
- Yanjing Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhuo Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yuli Geng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yufan Song
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Wenwen Ma
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Haoxu Dong
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Lijun Xu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Mingmin Zhang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Kunkun Song
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
50
|
Wu M, Liu H, Zhu Y, Wu P, Chen Y, Deng Z, Zhu X, Cai L. Bioinspired soft-hard combined system with mild photothermal therapeutic activity promotes diabetic bone defect healing via synergetic effects of immune activation and angiogenesis. Theranostics 2024; 14:4014-4057. [PMID: 38994032 PMCID: PMC11234279 DOI: 10.7150/thno.97335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
Background: The comprehensive management of diabetic bone defects remains a substantial clinical challenge due to the hostile regenerative microenvironment characterized by aggravated inflammation, excessive reactive oxygen species (ROS), bacterial infection, impaired angiogenesis, and unbalanced bone homeostasis. Thus, an advanced multifunctional therapeutic platform capable of simultaneously achieving immune regulation, bacterial elimination, and tissue regeneration is urgently designed for augmented bone regeneration under diabetic pathological milieu. Methods and Results: Herein, a photoactivated soft-hard combined scaffold system (PGCZ) was engineered by introducing polydopamine-modified zeolitic imidazolate framework-8-loaded double-network hydrogel (soft matrix component) into 3D-printed poly(ε-caprolactone) (PCL) scaffold (hard matrix component). The versatile PGCZ scaffold based on double-network hydrogel and 3D-printed PCL was thus prepared and features highly extracellular matrix-mimicking microstructure, suitable biodegradability and mechanical properties, and excellent photothermal performance, allowing long-term structural stability and mechanical support for bone regeneration. Under periodic near-infrared (NIR) irradiation, the localized photothermal effect of PGCZ triggers the on-demand release of Zn2+, which, together with repeated mild hyperthermia, collectively accelerates the proliferation and osteogenic differentiation of preosteoblasts and potently inhibits bacterial growth and biofilm formation. Additionally, the photoactivated PGCZ system also presents outstanding immunomodulatory and ROS scavenging capacities, which regulate M2 polarization of macrophages and drive functional cytokine secretion, thus leading to a pro-regenerative microenvironment in situ with enhanced vascularization. In vivo experiments further demonstrated that the PGCZ platform in conjunction with mild photothermal therapeutic activity remarkably attenuated the local inflammatory cascade, initiated endogenous stem cell recruitment and neovascularization, and orchestrated the osteoblast/osteoclast balance, ultimately accelerating diabetic bone regeneration. Conclusions: This work highlights the potential application of a photoactivated soft-hard combined system that provides long-term biophysical (mild photothermal stimulation) and biochemical (on-demand ion delivery) cues for accelerated healing of diabetic bone defects.
Collapse
Affiliation(s)
- Minhao Wu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan 430071 Hubei, China
| | - Huifan Liu
- Department of Anesthesiology, Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yufan Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan 430071 Hubei, China
| | - Ping Wu
- National Key laboratory of macromolecular drug development and manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yun Chen
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medicine Sciences), Wuhan University, Wuhan 430071, China
| | - Zhouming Deng
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan 430071 Hubei, China
| | - Xiaobin Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan 430071 Hubei, China
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 168 Donghu Street, Wuchang District, Wuhan 430071 Hubei, China
| |
Collapse
|