1
|
Chen Y, Chen L, Wu J, Xu X, Yang C, Zhang Y, Chen X, Lin K, Zhang S. Throw out an oligopeptide to catch a protein: Deep learning and natural language processing-screened tripeptide PSP promotes Osteolectin-mediated vascularized bone regeneration. Bioact Mater 2025; 46:37-54. [PMID: 39734571 PMCID: PMC11681832 DOI: 10.1016/j.bioactmat.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 09/26/2024] [Accepted: 11/06/2024] [Indexed: 12/31/2024] Open
Abstract
Angiogenesis is imperative for bone regeneration, yet the conventional cytokine therapies have been constrained by prohibitive costs and safety apprehensions. It is urgent to develop a safer and more efficient therapeutic alternative. Herein, utilizing the methodologies of Deep Learning (DL) and Natural Language Processing (NLP), we proposed a paradigm algorithm that amalgamates Word2vec with a TF-IDF variant, TF-IIDF, to deftly discern potential pro-angiogenic peptides from intrinsically disordered regions (IDRs) of 262 related proteins, where are fertile grounds for developing safer and highly promising bioactive peptides. After the evaluation of the candidate oligopeptides, one tripeptide, PSP, emerged as particularly notable for its exceptional ability to stimulate the vascularization of endothelial cells (ECs), enhance vascular-osteo communication, and then boost the osteogenic differentiation of bone marrow stem cells (BMSCs), evidenced in mouse critical-sized cranial model. Moreover, we found that PSP serves as a 'priming' agent, activating the body's innate ability to produce Osteolectin (Oln) - prompting ECs to release small extracellular vesicles (sEVs) enriched with Oln to facilitate bone formation. In summary, our study established a precise and efficient composite model of DL and NLP to screen bioactive peptides, opening an avenue for the development of various peptide-based therapeutic strategies applicable to a broader range of diseases.
Collapse
Affiliation(s)
- Yu Chen
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stom, Shanghai, 200011, China
| | - Long Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Jinyang Wu
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stom, Shanghai, 200011, China
| | - Xiaofeng Xu
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stom, Shanghai, 200011, China
| | - Chengshuai Yang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stom, Shanghai, 200011, China
| | - Yong Zhang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stom, Shanghai, 200011, China
| | - Xinrong Chen
- Academy for Engineering and Technology, Fudan University, Shanghai Key Laboratory of Medical Image Computing and Computer Assisted Intervention, Shanghai, 200000, China
| | - Kaili Lin
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stom, Shanghai, 200011, China
| | - Shilei Zhang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stom, Shanghai, 200011, China
| |
Collapse
|
2
|
Bai L, Li J, Li G, Zhou D, Su J, Liu C. Skeletal interoception and prospective application in biomaterials for bone regeneration. Bone Res 2025; 13:1. [PMID: 39743568 DOI: 10.1038/s41413-024-00378-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 01/04/2025] Open
Abstract
Accumulating research has shed light on the significance of skeletal interoception, in maintaining physiological and metabolic homeostasis related to bone health. This review provides a comprehensive analysis of how skeletal interoception influences bone homeostasis, delving into the complex interplay between the nervous system and skeletal system. One key focus of the review is the role of various factors such as prostaglandin E2 (PGE2) in skeletal health via skeletal interoception. It explores how nerves innervating the bone tissue communicate with the central nervous system to regulate bone remodeling, a process critical for maintaining bone strength and integrity. Additionally, the review highlights the advancements in biomaterials designed to utilize skeletal interoception for enhancing bone regeneration and treatment of bone disorders. These biomaterials, tailored to interact with the body's interoceptive pathways, are positioned at the forefront of innovative treatments for conditions like osteoporosis and fractures. They represent a convergence of bioengineering, neuroscience, and orthopedics, aiming to create more efficient and targeted therapies for bone-related disorders. In conclusion, the review underscores the importance of skeletal interoception in physiological regulation and its potential in developing more effective therapies for bone regeneration. It emphasizes the need for further research to fully understand the mechanisms of skeletal interoception and to harness its therapeutic potential fully.
Collapse
Affiliation(s)
- Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, China
| | - Jilong Li
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Guangfeng Li
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Dongyang Zhou
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Changsheng Liu
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
3
|
Tang K, Wang J, Pei X, Zhu Z, Liu J, Wan Q, Zhang X. Flexible coatings based on hydrogel to enhance the biointerface of biomedical implants. Adv Colloid Interface Sci 2025; 335:103358. [PMID: 39591835 DOI: 10.1016/j.cis.2024.103358] [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: 05/09/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
The use of biomedical implants in surgical techniques promotes the restoration of lost tissue or organ physiological functions in the body. The interface between different materials determines their interactions and ultimately affects the physicochemical properties of biomedical implants. After implantation, the biointerface plays a crucial role in determining the biocompatibility and functionality of biomedical implants. Surface modification of biomaterials by developing novel biomaterials like various flexible coatings to meet the requirements of biointerfaces, such as mechanical performance, compatibility safety, and biological activities, can improve material-biological interactions by maintaining its original volumetric characteristics. Hydrogels possess excellent plasticity, biodegradability, biocompatibility, and extracellular-matrix-like properties, making them widely used in the biomedical field. Moreover, due to their unique three-dimensional crosslinked hydrophilic network, hydrogels can encapsulate a variety of materials, such as small molecules, polymers, and particle. In recent years, it has been proved that coating biomedical implant materials with flexible hydrogels can optimize the biointerface and holds vast potential for implant surface modification. In this review, we first discussed the potential requirements of the biointerface on the surface of implantable materials in both in vitro and in vivo biological microenvironments. Based on these comprehensive reviews, we also introduced the potential applications of hydrogels in both in vitro and in vivo settings. Finally, this review focused on the challenges faced by the biointerface of implantable materials constructed based on hydrogels and proposed future approaches to inspire researchers with new ideas.
Collapse
Affiliation(s)
- Kun Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiang Pei
- 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 610041, Sichuan, China
| | - Zhou Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiayi Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xin Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
4
|
El-Ghannam A, Sultana F, Dréau D, Tiwari A, Yang IH, AlFotawi R, Knabe-Ducheyne C. Novel 3D printed bioactive SiC orthopedic screw promotes bone growth associated activities by macrophages, neurons, and osteoblasts. J Biomed Mater Res A 2025; 113:e37801. [PMID: 39319410 DOI: 10.1002/jbm.a.37801] [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: 06/15/2024] [Revised: 08/26/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024]
Abstract
Ceramic additive manufacturing currently relies on binders or high-energy lasers, each with limitations affecting final product quality and suitability for medical applications. To address these challenges, our laboratory has devised a surface activation technique for ceramic particles that eliminates the necessity for polymer binders or high-energy lasers in ceramic additive manufacturing. We utilized this method to 3D print bioactive SiC orthopedic screws and evaluated their properties. The study's findings reveal that chemical oxidation of SiC activated its surface, enabling 3D printing of orthopedic screws in a binder jet printer. Post-processing impregnation with NaOH and/or NH4OH strengthened the scaffold by promoting silica crystallization or partial conversion of silicon oxide into silicon nitride. The silica surface of the SiC 3D printed orthopedic screws facilitated osteoblast and neuron adhesion and extensive axon synthesis. The silicate ions released from the 3D printed SiC screws favorably modulated macrophage immune responses toward an M1 phenotype as indicated by the inhibition of TNFα secretions and of reactive oxygen species (ROS) expression along with the promotion of IL6R shedding. In contrast, under the same experimental conditions, Ti ions released from Ti6Al4V discs promoted macrophage TNFα secretion and ROS expression. In vivo tests demonstrated direct bone deposition on the SiC scaffold and a strong interfacial bond between the implanted SiC and bone. Immunostaining showed innervation, mineralization, and vascularization of the newly formed bone at the interface with SiC. Taken altogether, the 3D printed SiC orthopedic screws foster a favorable environment for wound healing and bone regeneration. The novel 3D printing method, based on ceramic surface activation represents a significant advancement in ceramic additive manufacturing and is applicable to a wide variety of materials.
Collapse
Affiliation(s)
- Ahmed El-Ghannam
- Department of Mechanical Engineering and Engineering Science, UNC Charlotte, Charlotte, North Carolina, USA
| | - Farjana Sultana
- Department of Mechanical Engineering and Engineering Science, UNC Charlotte, Charlotte, North Carolina, USA
| | - Didier Dréau
- Department of Biological Sciences, UNC Charlotte, Charlotte, North Carolina, USA
| | - Arjun Tiwari
- Department of Mechanical Engineering and Engineering Science, UNC Charlotte, Charlotte, North Carolina, USA
| | - In Hong Yang
- Department of Mechanical Engineering and Engineering Science, UNC Charlotte, Charlotte, North Carolina, USA
| | - Randa AlFotawi
- Department of Oral and Maxillofacial Surgery, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
5
|
Zhang H, Zhang P, Shen X, Han J, Wang H, Qin H, Wang B, Qian J, Udduttula A, Luo R, Zhao K, Wang Y, Chen Y. A cross-linked coating loaded with antimicrobial peptides for corrosion control, early antibacterial, and sequential osteogenic promotion on a magnesium alloy as orthopedic implants. Acta Biomater 2024:S1742-7061(24)00765-7. [PMID: 39716540 DOI: 10.1016/j.actbio.2024.12.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Magnesium (Mg)-based alloys have been recognized as desirable biodegradable materials for orthopedic implants. However, their clinical application has been limited by rapid degradation rates, insufficient antibacterial and osteogenic-promotion properties. Herein, a MgF2 priming layer was first constructed on AZ31 surface. Then, dopamine and polyphenols (EGCG) were cross-linked onto this AZ31-F surface to promote osteogenesis and further enhance corrosion protection, followed by chemical grafting of antimicrobial peptides (AMPs) via Michael-addition and Schiff-base reaction to confer antibacterial properties. In vitro electrochemical corrosion tests showed that icorr of AZ31-FE/AMPs (4.36×10-7 A/cm2) is two orders of magnitude lower than that of AZ31 (4.17×10-5 A/cm2). In vitro immersion degradation showed that AZ31-FE/AMPs exhibited the lowest hydrogen release (2.38 mL) after 400 h immersion with the lowest hydrogen evolution rate among them. Further, AZ31-FE/AMPs displayed inhibitory effects against S. aureus and E. coil in the initial stage and even after 7 days immersion in PBS (antibacterial rate > 85 %). AZ31-FE/AMPs promoted ALP secretion and calcium nodule formation in MC3T3-E1 cells. Transcriptome sequencing results indicated that osteogenic promotion mechanism of AZ31-FE/AMPs in MC3T3-E1 may involve the PI3K-Akt signalling pathway. Further, AZ31-FE/AMPs enhanced new bone formation when implanted in a rat femoral bone defect model. This coating strategy addresses initial antibacterial and later osteogenesis needs based on the corrosion control, which is crucial for the surface design of Mg-based implants. STATEMENT OF SIGNIFICANCE: It is critical for magnesium-based orthopedic implants to achieve sequential functions in the bone repair process while controlling an appropriate degradation rate. A MgF2 priming layer/phenolic-amine grafted AMPs (antimicrobial peptides) duplex coating was constructed on AZ31 surface in this study. The MgF2 layer provided a basic corrosion protection to magnesium substrate, and dopamine and polyphenols (EGCG) were then cross-linked to the MgF2 pretreated AZ31 to promote osteogenesis and enhance corrosion resistance, followed by chemical grafting of AMPs to confer antibacterial property. This strategy effectively meets the initial need for infection resistance and later osteogenic promotion on the basis of controlling the substrate corrosion rate, thus holding significant implications for the surface design of magnesium-based implants.
Collapse
Affiliation(s)
- Hao Zhang
- Institute of Vanadium and Titanium, Panzhihua University, Panzhihua, 617000, PR China
| | - Peng Zhang
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, PR China
| | - Xiaolong Shen
- College of Biology & Chemical Engineering (Agricultural College), Panzhihua University, Panzhihua 617000, PR China
| | - Jiaping Han
- Institute of Vanadium and Titanium, Panzhihua University, Panzhihua, 617000, PR China
| | - Haibo Wang
- Institute of Vanadium and Titanium, Panzhihua University, Panzhihua, 617000, PR China
| | - Haotian Qin
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, PR China
| | - Binbin Wang
- Institute of Vanadium and Titanium, Panzhihua University, Panzhihua, 617000, PR China
| | - Junyu Qian
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, PR China
| | - Anjaneyulu Udduttula
- Centre for Biomaterials, Vellore Institute of Technology (VIT), Cellular and Molecular Theranostics (CBCMT), Vellore, Tamil Nadu 632014, India
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, PR China
| | - Kexin Zhao
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, PR China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, PR China.
| | - Yingqi Chen
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, PR China.
| |
Collapse
|
6
|
Li B, Chen H, Hang R. Osseointegration-Related Exosomes for Surface Functionalization of Titanium Implants. Biomater Res 2024; 28:0124. [PMID: 39711824 PMCID: PMC11661649 DOI: 10.34133/bmr.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Despite that the clinical application of titanium-based implants has achieved great success, patients' own diseases and/or unhealthy lifestyle habits often lead to implant failure. Many studies have been carried out to modify titanium implants to promote osseointegration and implant success. Recent studies showed that exosomes, proactively secreted extracellular vesicles by mammalian cells, could selectively target and modulate the functions of recipient cells such as macrophages, nerve cells, endothelial cells, and bone marrow mesenchymal stem cells that are closely involved in implant osseointegration. Accordingly, using exosomes to functionalize titanium implants has been deemed as a novel and effective way to improve their osseointegration ability. Herein, recent advances pertaining to surface functionalization of titanium implants with exosomes are analyzed and discussed, with focus on the role of exosomes in regulating the functions of osseointegration-related cells, and their immobilization strategies as well as resultant impact on osseointegration ability.
Collapse
Affiliation(s)
- Boqiong Li
- Department of Materials Science and Engineering,
Jinzhong University, Jinzhong 030619, China
| | - Huanming Chen
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| |
Collapse
|
7
|
Jackett KN, Browne AT, Aber ER, Clements M, Kaplan RN. How the bone microenvironment shapes the pre-metastatic niche and metastasis. NATURE CANCER 2024; 5:1800-1814. [PMID: 39672975 DOI: 10.1038/s43018-024-00854-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/04/2024] [Indexed: 12/15/2024]
Abstract
The bone is a frequent metastatic site, with changes in the mineralized bone and the bone marrow milieu that can also prime other sites for metastasis by educating progenitor cells to support metastatic spread. Stromal and immune populations cooperatively maintain the organizationally complex bone niches and are dysregulated in the presence of a distant primary tumor and metastatic disease. Interrogating the bone niches that facilitate metastatic spread using innovative technologies holds the potential to aid in preventing metastasis in and mediated by the bone. Here, we review recent advances in bone niche biology and its adaptations in the context of cancer.
Collapse
Affiliation(s)
- Kailey N Jackett
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alice T Browne
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Etan R Aber
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Miranda Clements
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rosandra N Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
Li G, Gao F, Yang D, Lin L, Yu W, Tang J, Yang R, Jin M, Gu Y, Wang P, Lu E. ECM-mimicking composite hydrogel for accelerated vascularized bone regeneration. Bioact Mater 2024; 42:241-256. [PMID: 39285909 PMCID: PMC11404060 DOI: 10.1016/j.bioactmat.2024.08.035] [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: 05/15/2024] [Revised: 08/07/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Bioactive hydrogel materials have great potential for applications in bone tissue engineering. However, fabrication of functional hydrogels that mimic the natural bone extracellular matrix (ECM) remains a challenge, because they need to provide mechanical support and embody physiological cues for angiogenesis and osteogenesis. Inspired by the features of ECM, we constructed a dual-component composite hydrogel comprising interpenetrating polymer networks of gelatin methacryloyl (GelMA) and deoxyribonucleic acid (DNA). Within the composite hydrogel, the GelMA network serves as the backbone for mechanical and biological stability, whereas the DNA network realizes dynamic capabilities (e.g., stress relaxation), thereby promoting cell proliferation and osteogenic differentiation. Furthermore, functional aptamers (Apt19S and AptV) are readily attached to the DNA network to recruit bone marrow mesenchymal stem cells (BMSCs) and achieve sustained release of loaded vascular endothelial growth factor towards angiogenesis. Our results showed that the composite hydrogel could facilitate the adhesion of BMSCs, promote osteogenic differentiation by activating focal adhesion kinase (FAK)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/β-Catenin signaling pathway, and eventually enhance vascularized bone regeneration. This study shows that the multifunctional composite hydrogel of GelMA and DNA can successfully simulate the biological functions of natural bone ECM and has great potential for repairing bone defects.
Collapse
Affiliation(s)
- Guanglong Li
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Fei Gao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Donglei Yang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Lu Lin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Weijun Yu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Jiaqi Tang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Ruhan Yang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Min Jin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Yuting Gu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Pengfei Wang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| |
Collapse
|
9
|
Ghezzi B, Matera B, Meglioli M, Rossi F, Duraccio D, Faga MG, Zappettini A, Macaluso GM, Lumetti S. Composite PCL Scaffold With 70% β-TCP as Suitable Structure for Bone Replacement. Int Dent J 2024; 74:1220-1232. [PMID: 38614878 PMCID: PMC11551565 DOI: 10.1016/j.identj.2024.02.013] [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: 12/10/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 04/15/2024] Open
Abstract
OBJECTIVES The purpose of this work was to optimise printable polycaprolactone (PCL)/β-tricalcium phosphate (β-TCP) biomaterials with high percentages of β-TCP endowed with balanced mechanical characteristics to resemble human cancellous bone, presumably improving osteogenesis. METHODS PCL/β-TCP scaffolds were obtained from customised filaments for fused deposition modelling (FDM) 3D printing with increasing amounts of β-TCP. Samples mechanical features, surface topography and wettability were evaluated as well as cytocompatibility assays, cell adhesion and differentiation. RESULTS The parameters of the newly fabricated materila were optimal for PCL/β-TCP scaffold fabrication. Composite surfaces showed higher hydrophilicity compared with the controls, and their surface roughness sharply was higher, possibly due to the presence of β-TCP. The Young's modulus of the composites was significantly higher than that of pristine PCL, indicating that the intrinsic strength of β-TCP is beneficial for enhancing the elastic modulus of the composite biomaterials. All novel composite biomaterials supported greater cellular growth and stronger osteoblastic differentiation compared with the PCL control. CONCLUSIONS This project highlights the possibility to fabricat, through an FDM solvent-free approach, PCL/β-TCP scaffolds of up to 70 % concentrations of β-TCP. overcoming the current lmit of 60 % stated in the literature. The combination of 3D printing and customised biomaterials allowed production of highly personalised scaffolds with optimal mechanical and biological features resembling the natural structure and the composition of bone. This underlines the promise of such structures for innovative approaches for bone and periodontal regeneration.
Collapse
Affiliation(s)
- Benedetta Ghezzi
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy; Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Biagio Matera
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
| | - Matteo Meglioli
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy.
| | - Francesca Rossi
- Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Donatella Duraccio
- Istituto di Scienze e Tecnologie per l'Energia e la Mobilità Sostenibili, Consiglio Nazionale delle Ricerche, Torino, Italy
| | - Maria Giulia Faga
- Istituto di Scienze e Tecnologie per l'Energia e la Mobilità Sostenibili, Consiglio Nazionale delle Ricerche, Torino, Italy
| | - Andrea Zappettini
- Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Guido Maria Macaluso
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy; Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Simone Lumetti
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy; Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| |
Collapse
|
10
|
Wa Q, Luo Y, Tang Y, Song J, Zhang P, Linghu X, Lin S, Li G, Wang Y, Wen Z, Huang S, Xu W. Mesoporous bioactive glass-enhanced MSC-derived exosomes promote bone regeneration and immunomodulation in vitro and in vivo. J Orthop Translat 2024; 49:264-282. [PMID: 39524151 PMCID: PMC11550139 DOI: 10.1016/j.jot.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Background Exosomes produced by mesenchymal stem cells (MSCs) have vascular generative properties and are considered new effective candidates for the treatment of bone defects as alternatives to cell therapy. Improving the pro-regenerative function and efficacy of exosomes has been a popular research topic in the field of orthopaedics. Methods We prepared mesoporous bioactive glass (mBG) microspheres via the template method. The ionic products of mBGs used to treat MSCs were extracted, and the effects of exosomes secreted by MSCs on osteoblast (OB) and macrophage (MP) behaviour and bone defect repair were observed in vivo (Micro-CT, H&E, Masson, and immunofluorescence staining for BMP2, COL1, VEGF, CD31, CD163, and iNOS). Results The mBG spheres were successfully prepared, and the Exo-mBG were isolated and extracted. Compared with those in the blank and Exo-Con groups, the proliferation and osteogenic differentiation of OBs in the Exo-mBG group were significantly greater. For example, on Day 7, OPN gene expression in the Ctrl-Exo group was 3.97 and 2.83 times greater than that in the blank and Exo-mBG groups, respectively. In a cranial defect rat model, Exo-mBG promoted bone tissue healing and angiogenesis, increased M2 macrophage polarisation and inhibited M1 macrophage polarisation, as verified by micro-CT, H&E staining, Masson staining and immunofluorescence staining. These effects may be due to the combination of a higher silicon concentration and a higher calcium-to-phosphorus ratio in the mBG ionic products. Conclusion This study provides insights for the application of exosomes in cell-free therapy and a new scientific basis and technical approach for the utilisation of MSC-derived exosomes in bone defect repair. The translational potential of this article Our study demonstrated that exosomes produced by mBG-stimulated MSCs have excellent in vitro and in vivo bone-enabling and immunomodulatory functions and provides insights into the use of exosomes in clinical cell-free therapies.
Collapse
Affiliation(s)
- Qingde Wa
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China. Intersection of Xinlong Avenue and Xinpu Avenue, Honghuagang District, Zunyi, Guizhou, 563000, China
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, No.10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
| | - Yongxiang Luo
- Marshall Biomedical Engineering Laboratory, Shenzhen University, No. 3688 Nanhai Avenue, Nanshan District, Shenzhen, Guangdong, 518060, China
| | - Yubo Tang
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, China
| | - Jiaxiang Song
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China. Intersection of Xinlong Avenue and Xinpu Avenue, Honghuagang District, Zunyi, Guizhou, 563000, China
| | - Penghui Zhang
- Department of Orthopaedics, Seventh Affiliated Hospital of Sun Yat-sen University, No. 628, Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Xitao Linghu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China. Intersection of Xinlong Avenue and Xinpu Avenue, Honghuagang District, Zunyi, Guizhou, 563000, China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Yixiao Wang
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China, No. 98 Fenghuang North Road, Huichuan District, Zunyi City, Guizhou, 563002, China
| | - Zhenyu Wen
- Zunyi Medical University, No. 1 Campus, Xinpu New District, Zunyi City, Guizhou, 563000, China
| | - Shuai Huang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Haizhu District, Guangzhou, Guangdong, 510260, China
| | - Weikang Xu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, No.10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong, 510500, China
- Guangdong Chinese Medicine Intelligent Diagnosis and Treatment Engineering Technology Research Center, No. 10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
| |
Collapse
|
11
|
Zhang S, Huang L, Bian M, Xiao L, Zhou D, Tao Z, Zhao Z, Zhang J, Jiang LB, Li Y. Multifunctional Bone Regeneration Membrane with Flexibility, Electrical Stimulation Activity and Osteoinductive Activity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2405311. [PMID: 39148189 DOI: 10.1002/smll.202405311] [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: 06/28/2024] [Revised: 08/02/2024] [Indexed: 08/17/2024]
Abstract
The use of membrane-based guided bone regeneration techniques has great potential for single-stage reconstruction of critical-sized bone defects. Here, a multifunctional bone regeneration membrane combining flexible elasticity, electrical stimulation (ES) and osteoinductive activity is developed by in situ doping of MXene 2D nanomaterials with conductive functionality and β-TCP particles into a Poly(lactic acid-carbonate (PDT) composite nano-absorbable membrane (P/T/MXene) via electrostatic spinning technique. The composite membrane has good feasibility due to its temperature sensitivity, elastic memory capacity, coordinated degradation profile and easy preparation process. In vitro experiments showed the P/T/MXene membrane effectively promoted the recruitment and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) under ES and enhanced the angiogenic capacity of endothelial cells, which synergistically promoted bone regeneration through neovascularization. In addition, an in vivo rat model of cranial bone defects further confirmed the bone regeneration efficacy of the P/T/MXene membrane. In conclusion, the developed P/T/MXene membrane can effectively promote bone regeneration through their synergistic multifunctional effects, suggesting the membranes have great potential for guiding tissue regeneration and providing guidance for the biomaterials design.
Collapse
Affiliation(s)
- Shihao Zhang
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Lei Huang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lan Xiao
- School of Medicine and Dentistry, Griffith University, Australia, 4222, Australia
| | - Dong Zhou
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Ziwei Tao
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Zheng Zhao
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Jian Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li-Bo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yulin Li
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
12
|
Liu YD, Peng X, Chen HR, Liu XS, Peng LH. Nervonic acid as novel therapeutics initiates both neurogenesis and angiogenesis for comprehensive wound repair and healing. Front Pharmacol 2024; 15:1487183. [PMID: 39502529 PMCID: PMC11534657 DOI: 10.3389/fphar.2024.1487183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
Rapid tissue reconstruction in acute and chronic injuries are challengeable, the inefficient repair mainly due to the difficulty in simultaneous promoting the regeneration of peripheral nerves and vascular, which are closely related. Main clinical medication strategy of tissue repair depends on different cytokines to achieve nerves, blood vessels or granulation tissue regeneration, respectively. However, their effect is still limited to single aspect with biorisk exists upon long-time use. Herein, for the first time, we have demonstrated that NA isolated from Malania oleifera has potential to simultaneously promote both neurogenesis and angiogenesis in vitro and in vivo. First, NA was identified by NMR and FTIR structural characterization analysis. In a model of oxidative stress in neural cells induced by hydrogen peroxide, the cells viability of RSC96 and PC12 were protected from oxidative stress injury by NA. Similarly, based on the rat wound healing model, effective blood vessel formation and wound healing can be observed in tissue staining under NA treatment. In addition, according to the identification of nerve and vascular related markers in the wound tissue, the mechanism of NA promoting nerve regeneration lies in the upregulation of the secretion NGF, NF-200 and S100 protein, and NA treatment was also able to up-regulate VEGF and CD31 to directly promote angiogenesis during wound healing. This study provides an important candidate drug molecules for acute or chronic wound healing and nerve vascular synchronous regeneration.
Collapse
Affiliation(s)
- Yu-Da Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiao Peng
- Jinhua Institute, Zhejiang University, Jinhua, Zhejiang, China
| | - Hao-Ran Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xue-Song Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute, Zhejiang University, Jinhua, Zhejiang, China
| | - Li-Hua Peng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute, Zhejiang University, Jinhua, Zhejiang, China
| |
Collapse
|
13
|
Nokhbatolfoghahaei H, Baniameri S, Tabrizi R, Yousefi-Koma AA, Dehghan MM, Derakhshan S, Gharehdaghi N, Farzad-Mohajeri S, Behroozibakhsh M, Khojasteh A. Pre-vascularized porous gelatin-coated β-tricalcium phosphate scaffolds for bone regeneration: an in vivo and in vitro investigation. In Vitro Cell Dev Biol Anim 2024:10.1007/s11626-024-00973-5. [PMID: 39382735 DOI: 10.1007/s11626-024-00973-5] [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: 04/22/2024] [Accepted: 08/17/2024] [Indexed: 10/10/2024]
Abstract
Vascularization is vital in bone tissue engineering, supporting development, remodeling, and regeneration. Lack of vascularity leads to cell death, necessitating vascularization strategies. Angiogenesis, forming new blood vessels, provides crucial nutrients and oxygen. Pre-vascularized gelatin-coated β-tricalcium phosphate (G/β-TCP) scaffolds show promise in bone regeneration and vascularization. Our study evaluates G/β-TCP scaffolds' osteogenic and angiogenic potential in vitro and a canine model with vascular anastomosis. Channel-shaped G/β-TCP scaffolds were fabricated using foam casting and sintering of a calcium phosphate/silica slurry-coated polyurethane foam, then coated with cross-linked gelatin. Buccal fat pad-derived stem cells (BFPdSCs) were seeded onto scaffolds and assessed over time for adhesion, proliferation, and osteogenic capacity using scanning electron microscopy (SEM), 4,6-diamidino-2-phenylindole (DAPI) staining, Alamar blue, and alkaline phosphatase (ALP) assays. Scaffolds were implanted in a canine model to evaluate osteogenesis and angiogenesis by histology and CT scans at 12 wk. Our studies showed preliminary results for G/β-TCP scaffolds supporting angiogenesis and bone regeneration. In vitro analyses demonstrated excellent proliferation/viability, with BFPdSCs adhering and increasing on the scaffolds. ALP activity and protein levels increased, indicating osteogenic differentiation. Examination of tissue samples revealed granulation tissue with a well-developed vascular network, indicating successful angiogenesis and osteogenesis was further confirmed by a CT scan. In vivo, histology revealed scaffold resorption. However, scaffold placement beneath muscle tissue-restricted bone regeneration. Further optimization is needed for bone regeneration applications.
Collapse
Affiliation(s)
- Hanieh Nokhbatolfoghahaei
- Dental Research Center, School of Dentistry, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Baniameri
- Dental Research Center, School of Dentistry, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Research Assistance, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Tabrizi
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Ali Yousefi-Koma
- Dental Research Center, School of Dentistry, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Research Assistance, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Samira Derakhshan
- Oral and Maxillofacial Pathology Department, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Preclinical Imaging Group, Preclinical Core Facility, Tehran University of Medical Sciences, Tehran, Iran
| | - Niusha Gharehdaghi
- Dental Research Center, School of Dentistry, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Research Assistance, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Farzad-Mohajeri
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Marjan Behroozibakhsh
- Department of Dental Materials School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Khojasteh
- Dental Research Center, School of Dentistry, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Niu X, Xiao S, Huang R, Huang D, Aifantis KE, Yu H, Xue C, Yin L, Dunne N, Li X. ZIF-8-modified hydrogel sequentially delivers angiogenic and osteogenic growth factors to accelerate vascularized bone regeneration. J Control Release 2024; 374:154-170. [PMID: 39127448 DOI: 10.1016/j.jconrel.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
To realize high-quality vascularized bone regeneration, we developed a multifunctional hydrogel (SHPP-ZB) by incorporating BMP-2@ZIF-8/PEG-NH2 nanoparticles (NPs) into a sodium alginate/hydroxyapatite/polyvinyl alcohol hydrogel loaded with PDGF-BB, allowing for the sequential release of angiogenic and osteogenic growth factors (GFs) during bone repair. ZIF-8 served as a protective host for BMP-2 from degradation, ensuring high encapsulation efficiency and long-term bioactivity. The SHPP-ZB hydrogel exhibited enhanced mechanical strength and injectability, making it suitable for complex bone defects. It provided a swelling interface for tissue interlocking and the early release of Zn2+ and tannin acid (TA) to exert antioxidant and antibacterial effects, followed by the sequential release of angiogenic and osteogenic GFs to promote high-quality vascularized bone regeneration. In vitro experiments demonstrated the superior angiogenic and osteogenic properties of SHPP-ZB compared to other groups. In vivo experiments indicated that the sequential delivery of GFs via SHPP-ZB hydrogel could improve vascularized bone regeneration. Further, RNA sequencing analysis of regenerative bone tissue revealed that SHPP-ZB hydrogel promoted vascularized bone regeneration by regulating JUN, MAPK, Wnt, and calcium signaling pathways in vivo. This study presented a promising approach for efficient vascularized bone regeneration in large-scale bone defects.
Collapse
Affiliation(s)
- Xiaolian Niu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Shengzhao Xiao
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai 200011, China.
| | - Ruoyu Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-Biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Katerina E Aifantis
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Han Yu
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Chao Xue
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Lan Yin
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Nicholas Dunne
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| |
Collapse
|
15
|
Cho S, Lee KS, Lee K, Kim HS, Park S, Yu SE, Ha H, Baek S, Kim J, Kim H, Lee JY, Lee S, Sung HJ. Surface Crystal and Degradability of Shape Memory Scaffold Essentialize Osteochondral Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401989. [PMID: 38855993 DOI: 10.1002/smll.202401989] [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: 03/13/2024] [Revised: 05/27/2024] [Indexed: 06/11/2024]
Abstract
The minimally invasive deployment of scaffolds is a key safety factor for the regeneration of cartilage and bone defects. Osteogenesis relies primarily on cell-matrix interactions, whereas chondrogenesis relies on cell-cell aggregation. Bone matrix expansion requires osteoconductive scaffold degradation. However, chondrogenic cell aggregation is promoted on the repellent scaffold surface, and minimal scaffold degradation supports the avascular nature of cartilage regeneration. Here, a material satisfying these requirements for osteochondral regeneration is developed by integrating osteoconductive hydroxyapatite (HAp) with a chondroconductive shape memory polymer (SMP). The shape memory function-derived fixity and recovery of the scaffold enabled minimally invasive deployment and expansion to fill irregular defects. The crystalline phases on the SMP surface inhibited cell aggregation by suppressing water penetration and subsequent protein adsorption. However, HAp conjugation SMP (H-SMP) enhanced surface roughness and consequent cell-matrix interactions by limiting cell aggregation using crystal peaks. After mouse subcutaneous implantation, hydrolytic H-SMP accelerated scaffold degradation compared to that by the minimal degradation observed for SMP alone for two months. H-SMP and SMP are found to promote osteogenesis and chondrogenesis, respectively, in vitro and in vivo, including the regeneration of rat osteochondral defects using the binary scaffold form, suggesting that this material is promising for osteochondral regeneration.
Collapse
Affiliation(s)
- Sungwoo Cho
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kang Suk Lee
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- TMD LAB Co. Ltd., 6th Floor, 31, Gwangnaru-ro 8-gil, Seongdong-gu, Seoul, 04799, South Korea
| | - Kyubae Lee
- Department of Biomedical Materials, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, South Korea
| | - Hye-Seon Kim
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Suji Park
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seung Eun Yu
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyunsu Ha
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sewoom Baek
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jueun Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyunjae Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Ji Youn Lee
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sangmin Lee
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hak-Joon Sung
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- TMD LAB Co. Ltd., 6th Floor, 31, Gwangnaru-ro 8-gil, Seongdong-gu, Seoul, 04799, South Korea
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
16
|
Chen S, Zhou X, Li T, He C. Vascularization and Innervation for Bone Tissue Engineering. ACCOUNTS OF MATERIALS RESEARCH 2024; 5:1121-1133. [DOI: 10.1021/accountsmr.4c00165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Affiliation(s)
- 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
| | - 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
| | - Tao Li
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200092, 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
|
Leroux A, Roque M, Casas E, Leng J, Guibert C, L'Azou B, Oliveira H, Amédée J, Paiva Dos Santos B. The effect of CGRP and SP and the cell signaling dialogue between sensory neurons and endothelial cells. Biol Res 2024; 57:65. [PMID: 39261966 PMCID: PMC11389267 DOI: 10.1186/s40659-024-00538-6] [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: 05/09/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Increasing evidences demonstrate the role of sensory innervation in bone metabolism, remodeling and repair, however neurovascular coupling in bone is rarely studied. Using microfluidic devices as an indirect co-culture model to mimic in vitro the physiological scenario of innervation, our group demonstrated that sensory neurons (SNs) were able to regulate the extracellular matrix remodeling by endothelial cells (ECs), in particular through sensory neuropeptides, i.e. calcitonin gene-related peptide (CGRP) and substance P (SP). Nonetheless, still little is known about the cell signaling pathways and mechanism of action in neurovascular coupling. Here, in order to characterize the communication between SNs and ECs at molecular level, we evaluated the effect of SNs and the neuropeptides CGRP and SP on ECs. We focused on different pathways known to play a role on endothelial functions: calcium signaling, p38 and Erk1/2; the control of signal propagation through Cx43; and endothelial functions through the production of nitric oxide (NO). The effect of SNs was evaluated on ECs Ca2+ influx, the expression of Cx43, endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) production, p38, ERK1/2 as well as their phosphorylated forms. In addition, the role of CGRP and SP were either analyzed using respective antagonists in the co-culture model, or by adding directly on the ECs monocultures. We show that capsaicin-stimulated SNs induce increased Ca2+ influx in ECs. SNs stimulate the increase of NO production in ECs, probably involving a decrease in the inhibitory eNOS T495 phosphorylation site. The neuropeptide CGRP, produced by SNs, seems to be one of the mediators of this effect in ECs since NO production is decreased in the presence of CGRP antagonist in the co-culture of ECs and SNs, and increased when ECs are stimulated with synthetic CGRP. Taken together, our results suggest that SNs play an important role in the control of the endothelial cell functions through CGRP production and NO signaling pathway.
Collapse
Affiliation(s)
- Alice Leroux
- Univ. Bordeaux, INSERM, BIOTIS, Bordeaux, U1026, F-33000, France
| | - Micaela Roque
- Univ. Bordeaux, INSERM, BIOTIS, Bordeaux, U1026, F-33000, France
| | - Elina Casas
- Univ. Bordeaux, INSERM, BIOTIS, Bordeaux, U1026, F-33000, France
| | - Jacques Leng
- Univ. Bordeaux, CNRS, UMR 5258, Solvay, Pessac, LOF, F-33006, France
| | - Christelle Guibert
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Pessac, F-33604, France
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, F-33000, France
| | - Beatrice L'Azou
- Univ. Bordeaux, INSERM, BIOTIS, Bordeaux, U1026, F-33000, France
| | - Hugo Oliveira
- Univ. Bordeaux, INSERM, BIOTIS, Bordeaux, U1026, F-33000, France
| | - Joëlle Amédée
- Univ. Bordeaux, INSERM, BIOTIS, Bordeaux, U1026, F-33000, France
| | - Bruno Paiva Dos Santos
- Univ. Bordeaux, INSERM, BIOTIS, Bordeaux, U1026, F-33000, France.
- Univ. Paris Cité, URP2496-BRIO Pathologies Imagerie et Biothérapies Orofaciales, Montrouge, F-92120, France.
| |
Collapse
|
18
|
Zhang M, Huang Z, Wang X, Liu X, He W, Li Y, Wu D, Wu S. Personalized PLGA/BCL Scaffold with Hierarchical Porous Structure Resembling Periosteum-Bone Complex Enables Efficient Repair of Bone Defect. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401589. [PMID: 39018263 PMCID: PMC11425253 DOI: 10.1002/advs.202401589] [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: 02/14/2024] [Revised: 05/21/2024] [Indexed: 07/19/2024]
Abstract
Using bone regeneration scaffolds to repair craniomaxillofacial bone defects is a promising strategy. However, most bone regeneration scaffolds still exist some issues such as a lack of barrier structure, inability to precisely match bone defects, and necessity to incorporate biological components to enhance efficacy. Herein, inspired by a periosteum-bone complex, a class of multifunctional hierarchical porous poly(lactic-co-glycolic acid)/baicalein scaffolds is facilely prepared by the union of personalized negative mold technique and phase separation strategy and demonstrated to precisely fit intricate bone defect cavity. The dense up-surface of the scaffold can prevent soft tissue cell penetration, while the loose bottom-surface can promote protein adsorption, cell adhesion, and cell infiltration. The interior macropores of the scaffold and the loaded baicalein can synergistically promote cell differentiation, angiogenesis, and osteogenesis. These findings can open an appealing avenue for the development of personalized multifunctional hierarchical materials for bone repair.
Collapse
Affiliation(s)
- Mengqi Zhang
- Hospital of StomatologyGuanghua School of StomatologyGuangdong Provincial Key Laboratory of StomatologySun Yat‐sen UniversityGuangzhou510055P. R. China
| | - Zhike Huang
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080P. R. China
| | - Xun Wang
- Hospital of StomatologyGuanghua School of StomatologyGuangdong Provincial Key Laboratory of StomatologySun Yat‐sen UniversityGuangzhou510055P. R. China
| | - Xinyu Liu
- Hospital of StomatologyGuanghua School of StomatologyGuangdong Provincial Key Laboratory of StomatologySun Yat‐sen UniversityGuangzhou510055P. R. China
| | - Wenyi He
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of EducationSchool of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Yan Li
- Hospital of StomatologyGuanghua School of StomatologyGuangdong Provincial Key Laboratory of StomatologySun Yat‐sen UniversityGuangzhou510055P. R. China
| | - Dingcai Wu
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of EducationSchool of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Shuyi Wu
- Hospital of StomatologyGuanghua School of StomatologyGuangdong Provincial Key Laboratory of StomatologySun Yat‐sen UniversityGuangzhou510055P. R. China
| |
Collapse
|
19
|
de Mello Innocentini M, Fuzatto Bueno BR, Urbaś A, Morawska-Chochół A. Microstructural, Fluid Dynamic, and Mechanical Characterization of Zinc Oxide and Magnesium Chloride-Modified Hydrogel Scaffolds. ACS Biomater Sci Eng 2024; 10:4791-4801. [PMID: 39012256 PMCID: PMC11322906 DOI: 10.1021/acsbiomaterials.4c00286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/13/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Scaffolds for the filling and regeneration of osteochondral defects are a current challenge in the biomaterials field, and solutions with greater functionality are still being sought. The novel approach of this work was to obtain scaffolds with biologically active additives possessing microstructural, permeability, and mechanical properties, mimicking the complexity of natural cartilage. Four types of scaffolds with a gelatin/alginate matrix modified with hydroxyapatite were obtained, and the relationship between the modifiers and substrate properties was evaluated. They differed in the type of second modifier used, which was hydrated MgCl2 in two proportions, ZnO, and nanohydroxyapatite. The samples were obtained by freeze-drying by using two-stage freezing. Based on microstructural observations combined with X-ray microanalysis, the microstructure of the samples and the elemental content were assessed. Permeability and mechanical tests were also performed. The scaffolds exhibited a network of interconnected pores and complex microarchitecture, with lower porosity at the surface (15 ± 7 to 29 ± 6%) and higher porosity at the center (67 ± 8 to 75 ± 8%). The additives had varying effects on the pore sizes and permeabilities of the samples. ZnO yielded the most permeable scaffolds (5.92 × 10-11 m2), whereas nanohydroxyapatite yielded the scaffold with the lowest permeability (1.18 × 10-11 m2), values within the range reported for trabecular bone. The magnesium content had no statistically significant effect on the permeability. The best mechanical parameters were obtained for ZnO samples and those containing hydrated MgCl2. The scaffold's properties meet the criteria for filling osteochondral defects. The developed scaffolds follow a biomimetic approach in terms of hierarchical microarchitecture and mechanical parameters as well as chemical composition. The obtained composite materials have the potential as biomimetic scaffolds for the regeneration of osteochondral defects.
Collapse
Affiliation(s)
- Murilo
Daniel de Mello Innocentini
- Course
of Chemical Engineering, University of Ribeirão
Preto, Avenida Costabile Romano 2201, 14096-900 Ribeirão Preto, SP, Brazil
- Department
of Architecture and Civil Engineering, Centre for Regenerative Design
and Engineering for a Net Positive World (RENEW), University of Bath, Bath BA2 7AY, U.K.
| | - Bruno Ribeiro Fuzatto Bueno
- Course
of Chemical Engineering, University of Ribeirão
Preto, Avenida Costabile Romano 2201, 14096-900 Ribeirão Preto, SP, Brazil
| | - Agnieszka Urbaś
- Faculty
of Electrical Engineering, Automatics, Computer Science and Biomedical
Engineering, AGH University of Krakow, 30-059 Kraków, Poland
| | - Anna Morawska-Chochół
- Faculty
of Materials Science and Ceramics, Department of Biomaterials and
Composites, AGH University of Krakow, 30-059 Kraków, Poland
| |
Collapse
|
20
|
Xu W, Huang W, Cai X, Dang Z, Hao L, Wang L. Dexamethasone Long-Term Controlled Release from Injectable Dual-Network Hydrogels with Porous Microspheres Immunomodulation Promotes Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40581-40601. [PMID: 39074361 PMCID: PMC11311136 DOI: 10.1021/acsami.4c06661] [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: 04/23/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024]
Abstract
Long-lasting, controlled-release, and minimally invasive injectable platforms that provide a stable blood concentration to promote bone regeneration are less well developed. Using hexagonal mesoporous silica (HMS) loaded with dexamethasone (DEX) and poly(lactic-co-glycolic acid) (PLGA), we prepared porous DEX/HMS/PLGA microspheres (PDHP). In contrast to HMS/PLGA microspheres (HP), porous HMS/PLGA microspheres (PHP), DEX/PLGA microspheres (DP), and DEX/HMS/PLGA microspheres (DHP), PDHP showed notable immuno-coordinated osteogenic capabilities and were best at promoting bone mesenchymal stem cell proliferation and osteogenic differentiation. PDHP were combined with methacrylated silk (SilMA) and sodium alginate (SA) to form an injectable photocurable dual-network hydrogel platform that could continuously release the drug for more than 4 months. By adjusting the content of the microspheres in the hydrogel, a zero-order release hydrogel platform was obtained in vitro for 48 days. When the microsphere content was 1%, the hydrogel platform exhibited the best biocompatibility and osteogenic effects. The expression levels of the osteogenic gene alkaline phosphatases, BMP-2 and OPN were 10 to 15 times higher in the 1% group than in the 0% group, respectively. In addition, the 1% microsphere hydrogel strongly stimulated macrophage polarization to the M2 phenotype, establishing an immunological milieu that supports bone regrowth. The aforementioned outcomes were also observed in vivo. The most successful method for correcting cranial bone abnormalities in SD rats was to use a hydrogel called SilMA/SA containing 1% drug-loaded porous microspheres (PDHP/SS). The angiogenic and osteogenic effects of this treatment were also noticeably greater in the PDHP/SS group than in the control and blank groups. In addition, PDHP/SS polarized M2 macrophages and suppressed M1 macrophages in vivo, which reduced the local immune-inflammatory response, promoted angiogenesis, and cooperatively aided in situ bone healing. This work highlights the potential application of an advanced hydrogel platform for long-term, on-demand, controlled release for bone tissue engineering.
Collapse
Affiliation(s)
- Weikang Xu
- Institute
of Biological and Medical Engineering, Guangdong
Academy of Sciences, No. 10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou 510316, China
- National
Engineering Research Centre for Healthcare Devices, Guangdong Provincial
Key Laboratory of Medical Electronic Instruments and Materials, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou 510500, China
- Guangdong
Chinese Medicine Intelligent Diagnosis and Treatment Engineering Technology
Research Centre, No.
10 Shiliugang Road, Jianghai Avenue Central, Haizhu
District, Guangzhou 510316, China
| | - Weihua Huang
- Affiliated
Qingyuan Hospital, Guangzhou Medical University,
Qingyuan People’s Hospital, No. 35, Yinquan North Road, Qingcheng District, Qingyuan 511518, China
- National
Engineering Research Centre for Healthcare Devices, Guangdong Provincial
Key Laboratory of Medical Electronic Instruments and Materials, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou 510500, China
- Department
of Orthopaedic Surgery, the Second Affiliated Hospital of Guangzhou
Medical University, the Second Clinical
Medicine School of Guangzhou Medical University, No. 250 Changgang East Road, Haizhu
District, Guangzhou 510260, China
| | - Xiayu Cai
- Institute
of Biological and Medical Engineering, Guangdong
Academy of Sciences, No. 10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou 510316, China
- National
Engineering Research Centre for Healthcare Devices, Guangdong Provincial
Key Laboratory of Medical Electronic Instruments and Materials, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou 510500, China
| | - Zhaohui Dang
- Institute
of Biological and Medical Engineering, Guangdong
Academy of Sciences, No. 10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou 510316, China
- National
Engineering Research Centre for Healthcare Devices, Guangdong Provincial
Key Laboratory of Medical Electronic Instruments and Materials, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou 510500, China
- National
Engineering Research Centre for Human Tissue Restoration and Function
Reconstruction, South China University of
Technology, No. 381 Wushan Road, Guangzhou 510275, China
| | - Lijing Hao
- National
Engineering Research Centre for Human Tissue Restoration and Function
Reconstruction, South China University of
Technology, No. 381 Wushan Road, Guangzhou 510275, China
| | - Liyan Wang
- Department
of Stomatology, Foshan Women’s and Children’s Hospital, No. 11 Renmin Xi Road, Chancheng
District, Foshan 528000, China
| |
Collapse
|
21
|
Singhmar R, Son Y, Jo YJ, Zo S, Min BK, Sood A, Han SS. Fabrication of alginate composite hydrogel encapsulated retinoic acid and nano Se doped biphasic CaP to augment in situ mineralization and osteoimmunomodulation for bone regeneration. Int J Biol Macromol 2024; 275:133597. [PMID: 38960232 DOI: 10.1016/j.ijbiomac.2024.133597] [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: 04/10/2024] [Revised: 06/06/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Bone tissue engineering endows alternates to support bone defects/injuries that are circumscribed to undergo orchestrated process of remodeling on its own. In this regard, hydrogels have emerged as a promising platform that can confront irregular defects and encourage in situ bone repair. METHODS In this study, we aimed to develop a new approach for bone tissue regeneration by developing an alginate based composite hydrogel incorporating selenium doped biphasic calcium phosphate nanoparticles, and retinoic acid. The fabricated hydrogel was physiochemically evaluated for morphological, bonding, and mechanical behavior. Additionally, the biological response of the fabricated hydrogel was evaluated on MC3T3-E1 pre-osteoblast cells. RESULTS The developed composite hydrogel confers excellent biocompatibility, and osteoconductivity owing to the presence of alginate, and biphasic calcium phosphate, while selenium presents pro osteogenic, antioxidative, and immunomodulatory properties. The hydrogels exhibited highly porous microstructure, superior mechanical attributes, with enhanced calcification, and biomineralization abilities in vitro. SIGNIFICANCE By combining the osteoconductive properties of biphasic calcium phosphate with multifaceted benefits of selenium and retinoic acid, the fabricated composite hydrogel offers a potential transformation in the landscape of bone defect treatment. This strategy could direct a versatile and effective approach to tackle complex bone injuries/defects and present potential for clinical translation.
Collapse
Affiliation(s)
- Ritu Singhmar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Yumi Son
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Yoo Jung Jo
- Core Research Support Centre for Natural Products and Medical Materials, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Sunmi Zo
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Bong Ki Min
- Core Research Support Centre for Natural Products and Medical Materials, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Ankur Sood
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea; Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea; Core Research Support Centre for Natural Products and Medical Materials, 280 Daehak-ro, Gyeongsan 38541, South Korea; Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| |
Collapse
|
22
|
Ding Q, Liu W, Zhang S, Sun S, Yang J, Zhang L, Wang N, Ma S, Chai G, Shen L, Gao Y, Ding C, Liu X. Hydrogel loaded with thiolated chitosan modified taxifolin liposome promotes osteoblast proliferation and regulates Wnt signaling pathway to repair rat skull defects. Carbohydr Polym 2024; 336:122115. [PMID: 38670750 DOI: 10.1016/j.carbpol.2024.122115] [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: 01/04/2024] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024]
Abstract
To alleviate skull defects and enhance the biological activity of taxifolin, this study utilized the thin-film dispersion method to prepare paclitaxel liposomes (TL). Thiolated chitosan (CSSH)-modified TL (CTL) was synthesized through charge interactions. Injectable hydrogels (BLG) were then prepared as hydrogel scaffolds loaded with TAX (TG), TL (TLG), and CTL (CTLG) using a Schiff base reaction involving oxidized dextran and carboxymethyl chitosan. The study investigated the bone reparative properties of CTLG through molecular docking, western blot techniques, and transcriptome analysis. The particle sizes of CTL were measured at 248.90 ± 14.03 nm, respectively, with zeta potentials of +36.68 ± 5.43 mV, respectively. CTLG showed excellent antioxidant capacity in vitro. It also has a good inhibitory effect on Escherichia coli and Staphylococcus aureus, with inhibition rates of 93.88 ± 1.59 % and 88.56 ± 2.83 % respectively. The results of 5-ethynyl-2 '-deoxyuridine staining, alkaline phosphatase staining and alizarin red staining showed that CTLG also had the potential to promote the proliferation and differentiation of mouse embryonic osteoblasts (MC3T3-E1). The study revealed that CTLG enhances the expression of osteogenic proteins by regulating the Wnt signaling pathway, shedding light on the potential application of TAX and bone regeneration mechanisms.
Collapse
Affiliation(s)
- Qiteng Ding
- Jilin Agricultural University, Changchun 130118, China
| | - Wencong Liu
- School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543002, China
| | - Shuai Zhang
- Jilin Agricultural University, Changchun 130118, China
| | - Shuwen Sun
- Jilin Agricultural University, Changchun 130118, China
| | - Jiali Yang
- Jilin Agricultural University, Changchun 130118, China
| | - Lifeng Zhang
- Jilin Agricultural University, Changchun 130118, China
| | - Ning Wang
- Jilin Agricultural University, Changchun 130118, China
| | - Shuang Ma
- Jilin Agricultural University, Changchun 130118, China
| | - Guodong Chai
- Jilin Agricultural University, Changchun 130118, China
| | - Liqian Shen
- Jilin Jianwei Natural Biotechnology Co., Ltd., Linjiang 134600, China
| | - Yang Gao
- Jilin Jianwei Natural Biotechnology Co., Ltd., Linjiang 134600, China
| | - Chuanbo Ding
- Jilin Agricultural University, Changchun 130118, China; College of Traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; Jilin Aodong Yanbian Pharmaceutical Co., Ltd, Yanbian Korean Autonomous Prefecture 133000, China.
| | - Xinglong Liu
- College of Traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China.
| |
Collapse
|
23
|
Fois MG, van Griensven M, Giselbrecht S, Habibović P, Truckenmüller RK, Tahmasebi Birgani ZN. Mini-bones: miniaturized bone in vitro models. Trends Biotechnol 2024; 42:910-928. [PMID: 38493050 DOI: 10.1016/j.tibtech.2024.01.004] [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: 11/11/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 03/18/2024]
Abstract
In bone tissue engineering (TE) and regeneration, miniaturized, (sub)millimeter-sized bone models have become a popular trend since they bring about physiological biomimicry, precise orchestration of concurrent stimuli, and compatibility with high-throughput setups and high-content imaging. They also allow efficient use of cells, reagents, materials, and energy. In this review, we describe the state of the art of miniaturized in vitro bone models, or 'mini-bones', describing these models based on their characteristics of (multi)cellularity and engineered extracellular matrix (ECM), and elaborating on miniaturization approaches and fabrication techniques. We analyze the performance of 'mini-bone' models according to their applications for studying basic bone biology or as regeneration models, disease models, and screening platforms, and provide an outlook on future trends, challenges, and opportunities.
Collapse
Affiliation(s)
- Maria Gabriella Fois
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Martijn van Griensven
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Pamela Habibović
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Roman K Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands.
| | - Zeinab Niloofar Tahmasebi Birgani
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands.
| |
Collapse
|
24
|
Xiao F, Ye JH, Huang CX, Dai JH, Cheng KJ, Xu X, Deng LQ, You J, Liu YF. Gradient gyroid Ti6Al4V scaffolds with TiO 2 surface modification: Promising approach for large bone defect repair. BIOMATERIALS ADVANCES 2024; 161:213899. [PMID: 38772133 DOI: 10.1016/j.bioadv.2024.213899] [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: 02/19/2024] [Revised: 05/06/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024]
Abstract
Large bone defects, particularly those exceeding the critical size, present a clinical challenge due to the limited regenerative capacity of bone tissue. Traditional treatments like autografts and allografts are constrained by donor availability, immune rejection, and mechanical performance. This study aimed to develop an effective solution by designing gradient gyroid scaffolds with titania (TiO2) surface modification for the repair of large segmental bone defects. The scaffolds were engineered to balance mechanical strength with the necessary internal space to promote new bone formation and nutrient exchange. A gradient design of the scaffold was optimized through Finite Element Analysis (FEA) and Computational Fluid Dynamics (CFD) simulations to enhance fluid flow and cell adhesion. In vivo studies in rabbits demonstrated that the G@TiO2 scaffold, featuring a gradient structure and TiO2 surface modification, exhibited superior healing capabilities compared to the homogeneous structure and TiO2 surface modification (H@TiO2) and gradient structure (G) scaffolds. At 12 weeks post-operation, in a bone defect representing nearly 30 % of the total length of the radius, the implantation of the G@TiO2 scaffold achieved a 27 % bone volume to tissue volume (BV/TV) ratio, demonstrating excellent osseointegration. The TiO2 surface modification provided photothermal antibacterial effects, enhancing the scaffold's biocompatibility and potential for infection prevention. These findings suggest that the gradient gyroid scaffold with TiO2 surface modification is a promising candidate for treating large segmental bone defects, offering a combination of mechanical strength, bioactivity, and infection resistance.
Collapse
Affiliation(s)
- Fan Xiao
- College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Key Laboratory of Special Purpose Equipment and Advanced Processing Technology, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Collaborative Innovation Center of High-end Laser Manufacturing Equipment (National "2011 Plan"), Zhejiang University of Technology, Hangzhou 310023, People's Republic of China.
| | - Jun-Hui Ye
- College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Key Laboratory of Special Purpose Equipment and Advanced Processing Technology, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Collaborative Innovation Center of High-end Laser Manufacturing Equipment (National "2011 Plan"), Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Chen-Xiao Huang
- College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Key Laboratory of Special Purpose Equipment and Advanced Processing Technology, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Collaborative Innovation Center of High-end Laser Manufacturing Equipment (National "2011 Plan"), Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Jun-Hao Dai
- College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Key Laboratory of Special Purpose Equipment and Advanced Processing Technology, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Collaborative Innovation Center of High-end Laser Manufacturing Equipment (National "2011 Plan"), Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Kang-Jie Cheng
- College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Key Laboratory of Special Purpose Equipment and Advanced Processing Technology, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Collaborative Innovation Center of High-end Laser Manufacturing Equipment (National "2011 Plan"), Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Xu Xu
- Department of Stomatology, People's Hospital of Quzhou, Quzhou 324000, People's Republic of China
| | - Li-Quan Deng
- School/Hospital of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Jia You
- College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Key Laboratory of Special Purpose Equipment and Advanced Processing Technology, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Collaborative Innovation Center of High-end Laser Manufacturing Equipment (National "2011 Plan"), Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| | - Yun-Feng Liu
- College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Key Laboratory of Special Purpose Equipment and Advanced Processing Technology, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou 310023, People's Republic of China; Collaborative Innovation Center of High-end Laser Manufacturing Equipment (National "2011 Plan"), Zhejiang University of Technology, Hangzhou 310023, People's Republic of China
| |
Collapse
|
25
|
Mahmoudi N, Roque M, Paiva Dos Santos B, Oliveira H, Siadous R, Rey S, Garanger E, Lecommandoux S, Catros S, Garbay B, Amédée Vilamitjana J. An Elastin-Derived Composite Matrix for Enhanced Vascularized and Innervated Bone Tissue Reconstruction: From Material Development to Preclinical Evaluation. Adv Healthc Mater 2024; 13:e2303765. [PMID: 38651610 DOI: 10.1002/adhm.202303765] [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: 10/29/2023] [Revised: 03/28/2024] [Indexed: 04/25/2024]
Abstract
Despite progress in bone tissue engineering, reconstruction of large bone defects remains an important clinical challenge. Here, a biomaterial designed to recruit bone cells, endothelial cells, and neuronal fibers within the same matrix is developed, enabling bone tissue regeneration. The bioactive matrix is based on modified elastin-like polypeptides (ELPs) grafted with laminin-derived adhesion peptides IKVAV and YIGSR, and the SNA15 peptide for retention of hydroxyapatite (HA) particles. The composite matrix shows suitable porosity, interconnectivity, biocompatibility for endothelial cells, and the ability to support neurites outgrowth by sensory neurons. Subcutaneous implantation leads to the formation of osteoid tissue, characterized by the presence of bone cells, vascular networks, and neuronal structures, while minimizing inflammation. Using a rat femoral condyle defect model, longitudinal micro-CT analysis is performed, which demonstrates a significant increase in the volume of mineralized tissue when using the ELP-based matrix compared to empty defects and a commercially available control (Collapat). Furthermore, visible blood vessel networks and nerve fibers are observed within the lesions after a period of two weeks. By incorporating multiple key components that support cell growth, mineralization, and tissue integration, this ELP-based composite matrix provides a holistic and versatile solution to enhance bone tissue regeneration.
Collapse
Affiliation(s)
- Nadia Mahmoudi
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Micaela Roque
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Bruno Paiva Dos Santos
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Hugo Oliveira
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Robin Siadous
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Sylvie Rey
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | | | | | - Sylvain Catros
- CHU Bordeaux, Dentistry and Oral Health Department, Bordeaux, 33076, France
| | - Bertrand Garbay
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR, Pessac, 5629, France
| | | |
Collapse
|
26
|
Zheng L, Zhang R, Chen X, Luo Y, Du W, Zhu Y, Ruan YC, Xu J, Wang J, Qin L. Chronic kidney disease: a contraindication for using biodegradable magnesium or its alloys as potential orthopedic implants? Biomed Mater 2024; 19:045023. [PMID: 38815612 DOI: 10.1088/1748-605x/ad5241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
Magnesium (Mg) has gained widespread recognition as a potential revolutionary orthopedic biomaterial. However, whether the biodegradation of the Mg-based orthopedic implants would pose a risk to patients with chronic kidney disease (CKD) remains undetermined as the kidney is a key organ regulating mineral homeostasis. A rat CKD model was established by a 5/6 subtotal nephrectomy approach, followed by intramedullary implantation of three types of pins: stainless steel, high pure Mg with high corrosion resistance, and the Mg-Sr-Zn alloy with a fast degradation rate. The long-term biosafety of the biodegradable Mg or its alloys as orthopedic implants were systematically evaluated. During an experimental period of 12 weeks, the implantation did not result in a substantial rise of Mg ion concentration in serum or major organs such as hearts, livers, spleens, lungs, or kidneys. No pathological changes were observed in organs using various histological techniques. No significantly increased iNOS-positive cells or apoptotic cells in these organs were identified. The biodegradable Mg or its alloys as orthopedic implants did not pose an extra health risk to CKD rats at long-term follow-up, suggesting that these biodegradable orthopedic devices might be suitable for most target populations, including patients with CKD.
Collapse
Affiliation(s)
- Lizhen Zheng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Special Administrative Region of China, People's Republic of China
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, People's Republic of China
| | - Ri Zhang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, People's Republic of China
| | - Xin Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, People's Republic of China
| | - Ying Luo
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wanting Du
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China, People's Republic of China
| | - Yuwei Zhu
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, People's Republic of China
| | - Ye Chun Ruan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China, People's Republic of China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, People's Republic of China
| | - Jiali Wang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, People's Republic of China
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, People's Republic of China
- Hong Kong-Shenzhen Innovation and Technology Institute (Futian), The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, People's Republic of China
| |
Collapse
|
27
|
Zhu X, Wu S, Yang K, Wei W, Aziz Y, Yuan W, Miyatake H, Ito Y, Wei Z, Li J, Chen Y. Polydopamine-modified konjac glucomannan scaffold with sustained release of vascular endothelial growth factor to promote angiogenesis. Int J Biol Macromol 2024; 271:132333. [PMID: 38754686 DOI: 10.1016/j.ijbiomac.2024.132333] [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: 11/17/2023] [Revised: 04/24/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
The fabrication of scaffolds capable of the sustained release of the vascular endothelial growth factor (VEGF) to promote angiogenesis for a long time remains a challenge in tissue engineering. Here, we report a facile approach for effectively fabricating a bioactive scaffold that gradually releases VEGF to promote angiogenesis. The scaffold was fabricated by coating polydopamine (PDA) on a konjac glucomannan (KGM) scaffold, followed by the surface immobilization of VEGF with PDA. The resulting VEGF-PDA/KGM scaffold, with a porous and interconnected microstructure (392 μm pore size with 84.80 porosity), combined the features of long-term biodegradability (10 weeks with 51 % degradation rate), excellent biocompatibility, and sustained VEGF release for up to 21 days. The bioactive VEGF-PDA/KGM scaffold exhibited multiple angiogenic activities over time, as confirmed by in vivo and in vitro experiments. For example, the scaffold significantly promoted the attachment and proliferation of human umbilical vein endothelial cells and the formation of vascular tubes in vitro. Moreover, the in vivo results demonstrated the formation and maturation of blood vessels after subcutaneous implantation in rats for four weeks. This promising strategy is a feasible approach for producing bioactive materials that can induce angiogenesis in vivo. These findings provide a new avenue for designing and fabricating biocompatible and long-term biodegradable scaffolds for sustained VEGF release to facilitate angiogenesis.
Collapse
Affiliation(s)
- Xulong Zhu
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China; School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shuhan Wu
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Kuan Yang
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Wei Wei
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Yasir Aziz
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Wenjin Yuan
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Hideyuki Miyatake
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, Emergent Bioengineering Materials Research Team, RIKEN Center for emergent Matter Science, 2-1 Hirosawa, Wako, Saitama 3510198, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, Emergent Bioengineering Materials Research Team, RIKEN Center for emergent Matter Science, 2-1 Hirosawa, Wako, Saitama 3510198, Japan
| | - Zhao Wei
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jianhui Li
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China.
| | - Yongmei Chen
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| |
Collapse
|
28
|
Huang L, Cai P, Bian M, Yu J, Xiao L, Lu S, Wang J, Chen W, Han G, Xiang X, Liu X, Jiang L, Li Y, Zhang J. Injectable and high-strength PLGA/CPC loaded ALN/MgO bone cement for bone regeneration by facilitating osteogenesis and inhibiting osteoclastogenesis in osteoporotic bone defects. Mater Today Bio 2024; 26:101092. [PMID: 38873105 PMCID: PMC11169522 DOI: 10.1016/j.mtbio.2024.101092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/18/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Osteoporosis (OP) can result in slower bone regeneration than the normal condition due to the imbalance between osteogenesis and osteoclastogenesis, making osteoporotic bone defects healing a significant clinical challenge. Calcium phosphate cement (CPC) is a promising bone substitute material due to its good osteoinductive activity, however, the drawbacks such as fragility, slow degradation rate and incapability to control bone loss restrict its application in osteoporotic bone defects treatment. Currently, we developed the PLGA electrospun nanofiber sheets to carry alendronate (ALN) and magnesium oxide nanoparticle (nMgO) into CPC, therefore, to obtain a high-strength bone cement (C/AM-PL/C). The C/AM-PL/C bone cement had high mechanical strength, anti-washout ability, good injection performance and drug sustained release capacity. More importantly, the C/AM-PL/C cement promoted the osteogenic differentiation of bone marrow mesenchymal stem cells and neovascularization via the release of Mg2+ (from nMgO) and Ca2+ (during the degradation of CPC), and inhibited osteoclastogenesis via the release of ALN in vitro. Moreover, the injection of C/AM-PL/C cement significantly improved bone healing in an OP model with femur condyle defects in vivo. Altogether, the injectable C/AM-PL/C cement could facilitate osteoporotic bone regeneration, demonstrating its capacity as a promising candidate for treatment of osteoporotic bone defects.
Collapse
Affiliation(s)
- Lei Huang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Peihao Cai
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jieqin Yu
- Department of Orthopedic Surgery, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Lan Xiao
- School of Medicine and Dentistry, Griffith University, Gold COast, QLD, 4222, Australia
| | - Shunyi Lu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiayi Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weisin Chen
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guanjie Han
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xingdong Xiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin Liu
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Libo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yulin Li
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Jian Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
29
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [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: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
30
|
Xu Y, Xu C, Song H, Feng X, Ma L, Zhang X, Li G, Mu C, Tan L, Zhang Z, Liu Z, Luo Z, Yang C. Biomimetic bone-periosteum scaffold for spatiotemporal regulated innervated bone regeneration and therapy of osteosarcoma. J Nanobiotechnology 2024; 22:250. [PMID: 38750519 PMCID: PMC11094931 DOI: 10.1186/s12951-024-02430-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/20/2024] [Indexed: 05/19/2024] Open
Abstract
The complexity of repairing large segment defects and eradicating residual tumor cell puts the osteosarcoma clinical management challenging. Current biomaterial design often overlooks the crucial role of precisely regulating innervation in bone regeneration. Here, we develop a Germanium Selenium (GeSe) co-doped polylactic acid (PLA) nanofiber membrane-coated tricalcium phosphate bioceramic scaffold (TCP-PLA/GeSe) that mimics the bone-periosteum structure. This biomimetic scaffold offers a dual functionality, combining piezoelectric and photothermal conversion capabilities while remaining biodegradable. When subjected to ultrasound irradiation, the US-electric stimulation of TCP-PLA/GeSe enables spatiotemporal control of neurogenic differentiation. This feature supports early innervation during bone formation, promoting early neurogenic differentiation of Schwann cells (SCs) by increasing intracellular Ca2+ and subsequently activating the PI3K-Akt and Ras signaling pathways. The biomimetic scaffold also demonstrates exceptional osteogenic differentiation potential under ultrasound irradiation. In rabbit model of large segment bone defects, the TCP-PLA/GeSe demonstrates promoted osteogenesis and nerve fibre ingrowth. The combined attributes of high photothermal conversion capacity and the sustained release of anti-tumor selenium from the TCP-PLA/GeSe enable the synergistic eradication of osteosarcoma both in vitro and in vivo. This strategy provides new insights on designing advanced biomaterials of repairing large segment bone defect and osteosarcoma.
Collapse
Affiliation(s)
- Yan Xu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chao Xu
- College of Materials Science and Engineering, Wuhan Textile University, Wuhan, 430200, China
| | - Huan Song
- Otorhinolaryngology Head and Neck Surgery, Wuhan Fourth Hospital, Wuhan, Hubei, 430033, China
| | - Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaoguang Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Congpu Mu
- Center for High Pressure Science, State Key Laboratory of Metastable Materials Science and Technology, Yanshan University, Qinhuangdao, 066004, China
| | - Lei Tan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhengdong Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China.
- School of Clinical Medicine, Department of Orthopedics, Chengdu Medical College, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China.
| | - Zhongyuan Liu
- Center for High Pressure Science, State Key Laboratory of Metastable Materials Science and Technology, Yanshan University, Qinhuangdao, 066004, China
| | - Zhiqiang Luo
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
31
|
Li H, He Y, Chen X, Yang A, Lyu F, Dong Y. Exosomal miR-423-5p Derived from Cerebrospinal Fluid Pulsation Stress-Stimulated Osteoblasts Improves Angiogenesis of Endothelial Cells via DUSP8/ERK1/2 Signaling Pathway. Stem Cells Int 2024; 2024:5512423. [PMID: 38765936 PMCID: PMC11102110 DOI: 10.1155/2024/5512423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 05/22/2024] Open
Abstract
Exosomes secreted from osteoblasts (OBs) can regulate the angiogenesis of endothelial cells (ECs); however, whether cerebrospinal fluid pulsation (CSFP) stress, a special mechanical stimulation, can influence the cell's communication in the context of angiogenesis remains unknown. In this study, the effect of exosomes derived from CSFP stress-stimulated OBs on facilitating the angiogenesis of ECs was investigated. First, OBs were cultured in a CSFP bioreactor, and exosomes derived from OBs were isolated and identified. Cell Counting Kit 8 assay, transwell migration assay, wound healing migration assay, and tube formation assay were conducted to assess the effects of CSFP stress-stimulated OBs-derived exosomes (CSFP-Exos) on the angiogenesis of ECs. Then high-throughput RNA sequencing was used to determine the miRNA profiles of Non-CSFP stress-stimulated OBs-derived exosomes (NCSFP-Exos) and CSFP-Exos, and the luciferase reporter gene assay was performed to confirm the binging of miR-423-5p to DUSP8. In addition, the Matrigel plug assay was performed to explore whether exosomal miR-423-5p has the same effects in vivo. Our results suggested that CSFP-Exos can promote the angiogenesis of ECs, and miR-423-5p was enriched in CSFP-Exos. Moreover, miR-423-5p could promote the effect of angiogenesis via directly targeting dual-specificity phosphatase 8 (DUSP8), which inhibited the ERK1/2 signaling pathway. In conclusion, exosomal miR-423-5p derived from CSFP stress-stimulated OBs could promote the angiogenesis of ECs by the DUSP8/ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Hailong Li
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yiqun He
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xujun Chen
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Aolei Yang
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Feizhou Lyu
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Youhai Dong
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Li X, Cui Y, He X, Mao L. Hydrogel-Based Systems in Neuro-Vascularized Bone Regeneration: A Promising Therapeutic Strategy. Macromol Biosci 2024; 24:e2300484. [PMID: 38241425 DOI: 10.1002/mabi.202300484] [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: 10/25/2023] [Revised: 12/16/2023] [Indexed: 01/21/2024]
Abstract
Blood vessels and nerve fibers are distributed throughout the skeletal tissue, which enhance the development and function of each other and have an irreplaceable role in bone formation and remodeling. Despite significant progress in bone tissue engineering, the inadequacy of nerve-vascular network reconstruction remains a major limitation. This is partly due to the difficulty of integrating and regulating multiple tissue types with artificial materials. Thus, understanding the anatomy and underlying coupling mechanisms of blood vessels and nerve fibers within bone to further develop neuro-vascularized bone implant biomaterials is an extremely critical aspect in the field of bone regeneration. Hydrogels have good biocompatibility, controllable mechanical characteristics, and osteoconductive and osteoinductive properties, making them important candidates for research related to neuro-vascularized bone regeneration. This review reports the classification and physicochemical properties of hydrogels, with a focus on the application advantages and status of hydrogels for bone regeneration. The authors also highlight the effect of neurovascular coupling on bone repair and regeneration and the necessity of achieving neuro-vascularized bone regeneration. Finally, the recent progress and design strategies of hydrogel-based biomaterials for neuro-vascularized bone regeneration are discussed.
Collapse
Affiliation(s)
- Xiaojing Li
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200000, China
| | - Ya Cui
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200000, China
| | - Xiaoya He
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200000, China
| | - Lixia Mao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200000, China
| |
Collapse
|
33
|
Yu X, Ma H, Wang Y, Hao J, Chen L, Gelinsky M, Wu C. Assembled/Disassembled Modular Scaffolds for Multicellular Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308126. [PMID: 38533956 DOI: 10.1002/adma.202308126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 02/28/2024] [Indexed: 03/28/2024]
Abstract
The behavior of tissue resident cells can be influenced by the spatial arrangement of cellular interactions. Therefore, it is of significance to precisely control the spatial organization of various cells within multicellular constructs. It remains challenging to construct a versatile multicellular scaffold with ordered spatial organization of multiple cell types. Herein, a modular multicellular tissue engineering scaffold with ordered spatial distribution of different cell types is constructed by assembling varying cell-laden modules. Interestingly, the modular scaffolds can be disassembled into individual modules to evaluate the specific contribution of each cell type in the system. Through assembling cell-laden modules, the macrophage-mesenchymal stem cell (MSC), endothelial cell-MSC, and chondrocyte-MSC co-culture models are successfully established. The in vitro results indicate that the intercellular cross-talk can promote the proliferation and differentiation of each cell type in the system. Moreover, MSCs in the modular scaffolds may regulate the behavior of chondrocytes through the nuclear factor of activated T-Cells (NFAT) signaling pathway. Furthermore, the modular scaffolds loaded with co-cultured chondrocyte-MSC exhibit enhanced regeneration ability of osteochondral tissue, compared with other groups. Overall, this work offers a promising strategy to construct a multicellular tissue engineering scaffold for the systematic investigation of intercellular cross-talk and complex tissue engineering.
Collapse
Affiliation(s)
- Xiaopeng Yu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hongshi Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yufeng Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Jianxin Hao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Lei Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Michael Gelinsky
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Dresden University of Technology, Fetscherstr. 74, 01307, Dresden, Germany
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
34
|
Yang J, Chen Z, Gao C, Liu J, Liu K, Wang X, Pan X, Wang G, Sang H, Pan H, Liu W, Ruan C. A mechanical-assisted post-bioprinting strategy for challenging bone defects repair. Nat Commun 2024; 15:3565. [PMID: 38670999 PMCID: PMC11053166 DOI: 10.1038/s41467-024-48023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Bioprinting that can synchronously deposit cells and biomaterials has lent fresh impetus to the field of tissue regeneration. However, the unavoidable occurrence of cell damage during fabrication process and intrinsically poor mechanical stability of bioprinted cell-laden scaffolds severely restrict their utilization. As such, on basis of heart-inspired hollow hydrogel-based scaffolds (HHSs), a mechanical-assisted post-bioprinting strategy is proposed to load cells into HHSs in a rapid, uniform, precise and friendly manner. HHSs show mechanical responsiveness to load cells within 4 s, a 13-fold increase in cell number, and partitioned loading of two types of cells compared with those under static conditions. As a proof of concept, HHSs with the loading cells show an enhanced regenerative capability in repair of the critical-sized segmental and osteoporotic bone defects in vivo. We expect that this post-bioprinting strategy can provide a universal, efficient, and promising way to promote cell-based regenerative therapy.
Collapse
Affiliation(s)
- Jirong Yang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhigang Chen
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chongjian Gao
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Juan Liu
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kaizheng Liu
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiao Wang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
| | - Xiaoling Pan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
| | - Guocheng Wang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongxun Sang
- Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
| | - Haobo Pan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Changshun Ruan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
35
|
Xu Q, Bai Y, Li S, Hou W, Hao Y, Yang R, Li X, Zhang X. Enhancing osteogenesis and angiogenesis functions for Ti-24Nb-4Zr-8Sn scaffolds with methacrylated gelatin and deferoxamine. Front Bioeng Biotechnol 2024; 12:1372636. [PMID: 38707506 PMCID: PMC11066197 DOI: 10.3389/fbioe.2024.1372636] [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: 01/18/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
Repair of large bone defects remains challenge for orthopedic clinical treatment. Porous titanium alloys have been widely fabricated by the additive manufacturing, which possess the elastic modulus close to that of human cortical bone, good osteoconductivity and osteointegration. However, insufficient bone regeneration and vascularization inside the porous titanium scaffolds severely limit their capability for repair of large-size bone defects. Therefore, it is crucially important to improve the osteogenic function and vascularization of the titanium scaffolds. Herein, methacrylated gelatin (GelMA) were incorporated with the porous Ti-24Nb-4Zr-8Sn (Ti2448) scaffolds prepared by the electron beam melting (EBM) method (Ti2448-GelMA). Besides, the deferoxamine (DFO) as an angiogenic agent was doped into the Ti2448-GelMA scaffold (Ti2448-GelMA/DFO), in order to promote vascularization. The results indicate that GelMA can fully infiltrate into the pores of Ti2448 scaffolds with porous cross-linked network (average pore size: 120.2 ± 25.1 μm). Ti2448-GelMA scaffolds facilitated the differentiation of MC3T3-E1 cells by promoting the ALP expression and mineralization, with the amount of calcium contents ∼2.5 times at day 14, compared with the Ti2448 scaffolds. Impressively, the number of vascular meshes for the Ti2448-GelMA/DFO group (∼7.2/mm2) was significantly higher than the control group (∼5.3/mm2) after cultivation for 9 h, demonstrating the excellent angiogenesis ability. The Ti2448-GelMA/DFO scaffolds also exhibited sustained release of DFO, with a cumulative release of 82.3% after 28 days. Therefore, Ti2448-GelMA/DFO scaffolds likely provide a new strategy to improve the osteogenesis and angiogenesis for repair of large bone defects.
Collapse
Affiliation(s)
- Qian Xu
- Department of Materials Physics and Chemistry, School of Materials Science and Engineering, Key Laboratory for Anisotropy and Texture of Materials, Ministry of Education, Northeastern University, Shenyang, Liaoning, China
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
| | - Yun Bai
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Shujun Li
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Wentao Hou
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Yulin Hao
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Rui Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaowu Li
- Department of Materials Physics and Chemistry, School of Materials Science and Engineering, Key Laboratory for Anisotropy and Texture of Materials, Ministry of Education, Northeastern University, Shenyang, Liaoning, China
| | - Xing Zhang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
36
|
Zhou P, Liu T, Liu W, Sun L, Kang H, Liu K, Luo P, Wang Y, Luo L, Dai H. An Antibacterial Bionic Periosteum with Angiogenesis-Neurogenesis Coupling Effect for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38623938 DOI: 10.1021/acsami.4c01206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The periosteum, rich in neurovascular networks, bone progenitor cells, and stem cells, is vital for bone repair. Current artificial periosteal materials face challenges in mechanical strength, bacterial infection, and promoting osteogenic differentiation and angiogenesis. To address these issues, we adjusted the electrospinning ratio of poly-ε-caprolactone and chitosan and incorporated Zn doping whitlockite with polydopamine coating into a nanofiber membrane. After a series of characterizations, optimal results were achieved with a poly-ε-caprolactone: chitosan ratio of 8:1 and 5% nanoparticle content. In vitro cell experiments and in vivo calvarial defect models, the sustained release of Mg2+ and Ca2+ promoted vascularization and new bone formation, respectively, while the release of Zn2+ was conducive to antibacterial and cooperated with Mg2+ to promote neurovascularization. Consequently, this antibacterial bionic periosteum with an angiogenesis-neurogenesis coupling effect demonstrates a promising potential for bone repair applications.
Collapse
Affiliation(s)
- Peiqian Zhou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Tuozhou Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 511442, China
| | - Wenbin Liu
- Department of Orthopaedics, The Third Xiangya Hospital Central South University, 138 Tongzipo Road, Changsha, Hunan 410008, China
| | - Lingshun Sun
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Haifei Kang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Kun Liu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Peiyuan Luo
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Youfa Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Ling Luo
- Department of Orthopaedics, The Third Xiangya Hospital Central South University, 138 Tongzipo Road, Changsha, Hunan 410008, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
- National Energy Key Laboratory for New Hydrogen-Ammonia Energy Technologies, Foshan Xianhu Laboratory, Foshan 528200, China
| |
Collapse
|
37
|
Damiati LA, El Soury M. Bone-nerve crosstalk: a new state for neuralizing bone tissue engineering-A mini review. Front Med (Lausanne) 2024; 11:1386683. [PMID: 38690172 PMCID: PMC11059066 DOI: 10.3389/fmed.2024.1386683] [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: 02/15/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
Neuro bone tissue engineering is a multidisciplinary field that combines both principles of neurobiology and bone tissue engineering to develop innovative strategies for repairing and regenerating injured bone tissues. Despite the fact that regeneration and development are considered two distinct biological processes, yet regeneration can be considered the reactivation of development in later life stages to restore missing tissues. It is noteworthy that the regeneration capabilities are distinct and vary from one organism to another (teleost fishes, hydra, humans), or even in the same organism can vary dependent on the injured tissue itself (Human central nervous system vs. peripheral nervous system). The skeletal tissue is highly innervated, peripheral nervous system plays a role in conveying the signals and connecting the central nervous system with the peripheral organs, moreover it has been shown that they play an important role in tissue regeneration. Their regeneration role is conveyed by the different cells' resident in it and in its endoneurium (fibroblasts, microphages, vasculature associated cells, and Schwann cells) these cells secrete various growth factors (NGF, BDNF, GDNF, NT-3, and bFGF) that contribute to the regenerative phenotype. The peripheral nervous system and central nervous system synchronize together in regulating bone homeostasis and regeneration through neurogenic factors and neural circuits. Receptors of important central nervous system peptides such as Serotonin, Leptin, Semaphorins, and BDNF are expressed in bone tissue playing a role in bone homeostasis, metabolism and regeneration. This review will highlight the crosstalk between peripheral nerves and bone in the developmental stages as well as in regeneration and different neuro-bone tissue engineering strategies for repairing severe bone injuries.
Collapse
Affiliation(s)
- Laila A. Damiati
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Marwa El Soury
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| |
Collapse
|
38
|
Cao X, Chen Y, Zhang C, Mao Z, Zhang J, Ma T, Tian W, Kong X, Li H, Rao S, Yang K. Heterogeneous nucleation induced A. pernyi/B. mori silk fibroin coatings on AZ31 biometals with enhanced corrosion resistance, adhesion and biocompatibility. Int J Biol Macromol 2024; 264:130524. [PMID: 38442832 DOI: 10.1016/j.ijbiomac.2024.130524] [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: 01/02/2024] [Revised: 02/10/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024]
Abstract
Silk fibroin coatings on biomedical magnesium alloys have garnered significant attention due to their enhanced corrosion resistance and biocompatibility. However, the utilization of wild A. pernyi silk fibroin, known for its RGD sequence that facilitates tissue regeneration, presents a challenge for corrosion-resistant coatings on magnesium alloys due to its weak adhesion and high dissolution rate. In this study, we employed hexafluoroisopropanol as a solvent to blend A. pernyi silk fibroin with B. mori silk fibroin. The resulting blended fibroin coating at a 3:7 mass ratio exhibited a heterogeneous nucleation effect, enhancing β-sheet content (32.3 %) and crystallinity (28.6 %). This improved β-sheet promoted the "labyrinth effect" with an Icorr of 2.15 × 10-6 A cm-2, resulting in significantly improved corrosion resistance, which is two orders of magnitude lower than that of pure magnesium alloy. Meanwhile, the increased content of exposed serine in zigzag β-sheet contributes to a higher adhesion strength. Cell cytotoxicity evaluation confirmed the enhanced cell adhesion and bioactivity. This work provides a facile approach for wild A. pernyi silk fibroin coatings on magnesium alloys with enhanced corrosion resistance, adhesion and biocompatibility.
Collapse
Affiliation(s)
- Xinru Cao
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Yanning Chen
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China.
| | - Chen Zhang
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Zhinan Mao
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Jingwu Zhang
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Tingji Ma
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Wenhan Tian
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science and Engineering, Beihang University, Beijing 100191, China
| | - Xiangsheng Kong
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Haotong Li
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Sixian Rao
- School of Mechanical Engineering, Anhui University of Technology, Maanshan 243002, China.
| | - Kang Yang
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China; School of Mechanical Engineering, Anhui University of Technology, Maanshan 243002, China.
| |
Collapse
|
39
|
Liu J, Zhou Z, Hou M, Xia X, Liu Y, Zhao Z, Wu Y, Deng Y, Zhang Y, He F, Xu Y, Zhu X. Capturing cerium ions via hydrogel microspheres promotes vascularization for bone regeneration. Mater Today Bio 2024; 25:100956. [PMID: 38322657 PMCID: PMC10844749 DOI: 10.1016/j.mtbio.2024.100956] [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: 10/27/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
The rational design of multifunctional biomaterials with hierarchical porous structure and on-demand biological activity is of great consequence for bone tissue engineering (BTE) in the contemporary world. The advanced combination of trace element cerium ions (Ce3+) with bone repair materials makes the composite material capable of promoting angiogenesis and enhancing osteoblast activity. Herein, a living and phosphorylated injectable porous hydrogel microsphere (P-GelMA-Ce@BMSCs) is constructed by microfluidic technology and coordination reaction with metal ion ligands while loaded with exogenous BMSCs. Exogenous stem cells can adhere to and proliferate on hydrogel microspheres, thus promoting cell-extracellular matrix (ECM) and cell-cell interactions. The active ingredient Ce3+ promotes the proliferation, osteogenic differentiation of rat BMSCs, and angiogenesis of endotheliocytes by promoting mineral deposition, osteogenic gene expression, and VEGF secretion. The enhancement of osteogenesis and improvement of angiogenesis of the P-GelMA-Ce scaffold is mainly associated with the activation of the Wnt/β-catenin pathway. This study could provide novel and meaningful insights for treating bone defects with biofunctional materials on the basis of metal ions.
Collapse
Affiliation(s)
- Junlin Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Zhangzhe Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Xiaowei Xia
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Yang Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Zhijian Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Yubin Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Yaoge Deng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Yijian Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Yong Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| |
Collapse
|
40
|
Zhang H, Qin C, Shi Z, Xue J, Hao J, Huang J, Du L, Lu H, Wu C. Bioprinting of inorganic-biomaterial/neural-stem-cell constructs for multiple tissue regeneration and functional recovery. Natl Sci Rev 2024; 11:nwae035. [PMID: 38463933 PMCID: PMC10924618 DOI: 10.1093/nsr/nwae035] [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: 11/13/2023] [Revised: 01/14/2024] [Accepted: 01/24/2024] [Indexed: 03/12/2024] Open
Abstract
Tissue regeneration is a complicated process that relies on the coordinated effort of the nervous, vascular and immune systems. While the nervous system plays a crucial role in tissue regeneration, current tissue engineering approaches mainly focus on restoring the function of injury-related cells, neglecting the guidance provided by nerves. This has led to unsatisfactory therapeutic outcomes. Herein, we propose a new generation of engineered neural constructs from the perspective of neural induction, which offers a versatile platform for promoting multiple tissue regeneration. Specifically, neural constructs consist of inorganic biomaterials and neural stem cells (NSCs), where the inorganic biomaterials endows NSCs with enhanced biological activities including proliferation and neural differentiation. Through animal experiments, we show the effectiveness of neural constructs in repairing central nervous system injuries with function recovery. More importantly, neural constructs also stimulate osteogenesis, angiogenesis and neuromuscular junction formation, thus promoting the regeneration of bone and skeletal muscle, exhibiting its versatile therapeutic performance. These findings suggest that the inorganic-biomaterial/NSC-based neural platform represents a promising avenue for inducing the regeneration and function recovery of varying tissues and organs.
Collapse
Affiliation(s)
- Hongjian Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Qin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Zhe Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Jianmin Xue
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Jianxin Hao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinzhou Huang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Du
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongxu Lu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
41
|
Yang J, Fatima K, Zhou X, He C. Meticulously engineered three-dimensional-printed scaffold with microarchitecture and controlled peptide release for enhanced bone regeneration. BIOMATERIALS TRANSLATIONAL 2024; 5:69-83. [PMID: 39220663 PMCID: PMC11362348 DOI: 10.12336/biomatertransl.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/08/2024] [Accepted: 02/29/2024] [Indexed: 09/04/2024]
Abstract
The repair of large load-bearing bone defects requires superior mechanical strength, a feat that a single hydrogel scaffold cannot achieve. The objective is to seamlessly integrate optimal microarchitecture, mechanical robustness, vascularisation, and osteoinductive biological responses to effectively address these critical load-bearing bone defects. To confront this challenge, three-dimensional (3D) printing technology was employed to prepare a polycaprolactone (PCL)-based integrated scaffold. Within the voids of 3D printed PCL scaffold, a methacrylate gelatin (GelMA)/methacrylated silk fibroin (SFMA) composite hydrogel incorporated with parathyroid hormone (PTH) peptide-loaded mesoporous silica nanoparticles (PTH@MSNs) was embedded, evolving into a porous PTH@MSNs/GelMA/SFMA/PCL (PM@GS/PCL) scaffold. The feasibility of fabricating this functional scaffold with a customised hierarchical structure was confirmed through meticulous chemical and physical characterisation. Compression testing unveiled an impressive strength of 17.81 ± 0.83 MPa for the composite scaffold. Additionally, in vitro angiogenesis potential of PM@GS/PCL scaffold was evaluated through Transwell and tube formation assays using human umbilical vein endothelium, revealing the superior cell migration and tube network formation. The alizarin red and alkaline phosphatase staining assays using bone marrow-derived mesenchymal stem cells clearly illustrated robust osteogenic differentiation properties within this scaffold. Furthermore, the bone repair potential of the scaffold was investigated on a rat femoral defect model using micro-computed tomography and histological examination, demonstrating enhanced osteogenic and angiogenic performance. This study presents a promising strategy for fabricating a microenvironment-matched composite scaffold for bone tissue engineering, providing a potential solution for effective bone defect repair.
Collapse
Affiliation(s)
- Jin Yang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine; College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Kanwal Fatima
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine; College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Xiaojun Zhou
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine; College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Chuanglong He
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine; College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| |
Collapse
|
42
|
Loukelis K, Koutsomarkos N, Mikos AG, Chatzinikolaidou M. Advances in 3D bioprinting for regenerative medicine applications. Regen Biomater 2024; 11:rbae033. [PMID: 38845855 PMCID: PMC11153344 DOI: 10.1093/rb/rbae033] [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: 12/03/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 06/09/2024] Open
Abstract
Biofabrication techniques allow for the construction of biocompatible and biofunctional structures composed from biomaterials, cells and biomolecules. Bioprinting is an emerging 3D printing method which utilizes biomaterial-based mixtures with cells and other biological constituents into printable suspensions known as bioinks. Coupled with automated design protocols and based on different modes for droplet deposition, 3D bioprinters are able to fabricate hydrogel-based objects with specific architecture and geometrical properties, providing the necessary environment that promotes cell growth and directs cell differentiation towards application-related lineages. For the preparation of such bioinks, various water-soluble biomaterials have been employed, including natural and synthetic biopolymers, and inorganic materials. Bioprinted constructs are considered to be one of the most promising avenues in regenerative medicine due to their native organ biomimicry. For a successful application, the bioprinted constructs should meet particular criteria such as optimal biological response, mechanical properties similar to the target tissue, high levels of reproducibility and printing fidelity, but also increased upscaling capability. In this review, we highlight the most recent advances in bioprinting, focusing on the regeneration of various tissues including bone, cartilage, cardiovascular, neural, skin and other organs such as liver, kidney, pancreas and lungs. We discuss the rapidly developing co-culture bioprinting systems used to resemble the complexity of tissues and organs and the crosstalk between various cell populations towards regeneration. Moreover, we report on the basic physical principles governing 3D bioprinting, and the ideal bioink properties based on the biomaterials' regenerative potential. We examine and critically discuss the present status of 3D bioprinting regarding its applicability and current limitations that need to be overcome to establish it at the forefront of artificial organ production and transplantation.
Collapse
Affiliation(s)
- Konstantinos Loukelis
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Nikos Koutsomarkos
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion 70013, Greece
| |
Collapse
|
43
|
Lian M, Qiao Z, Qiao S, Zhang X, Lin J, Xu R, Zhu N, Tang T, Huang Z, Jiang W, Shi J, Hao Y, Lai H, Dai K. Nerve Growth Factor-Preconditioned Mesenchymal Stem Cell-Derived Exosome-Functionalized 3D-Printed Hierarchical Porous Scaffolds with Neuro-Promotive Properties for Enhancing Innervated Bone Regeneration. ACS NANO 2024; 18:7504-7520. [PMID: 38412232 DOI: 10.1021/acsnano.3c11890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
The essential role of the neural network in enhancing bone regeneration has often been overlooked in biomaterial design, leading to delayed or compromised bone healing. Engineered mesenchymal stem cells (MSCs)-derived exosomes are becoming increasingly recognized as potent cell-free agents for manipulating cellular behavior and improving therapeutic effectiveness. Herein, MSCs are stimulated with nerve growth factor (NGF) to regulate exosomal cargoes to improve neuro-promotive potential and facilitate innervated bone regeneration. In vitro cell experiments showed that the NGF-stimulated MSCs-derived exosomes (N-Exos) obviously improved the cellular function and neurotrophic effects of the neural cells, and consequently, the osteogenic potential of the osteo-reparative cells. Bioinformatic analysis by miRNA sequencing and pathway enrichment revealed that the beneficial effects of N-Exos may partly be ascribed to the NGF-elicited multicomponent exosomal miRNAs and the subsequent regulation and activation of the MAPK and PI3K-Akt signaling pathways. On this basis, N-Exos were delivered on the micropores of the 3D-printed hierarchical porous scaffold to accomplish the sustained release profile and extended bioavailability. In a rat model with a distal femoral defect, the N-Exos-functionalized hierarchical porous scaffold significantly induced neurovascular structure formation and innervated bone regeneration. This study provided a feasible strategy to modulate the functional cargoes of MSCs-derived exosomes to acquire desirable neuro-promotive and osteogenic potential. Furthermore, the developed N-Exos-functionalized hierarchical porous scaffold may represent a promising neurovascular-promotive bone reparative scaffold for clinical translation.
Collapse
Affiliation(s)
- Meifei Lian
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Zhiguang Qiao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Shichong Qiao
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Xing Zhang
- State Key Laboratory of Mechanical Systems and Vibration, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jieming Lin
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Ruida Xu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Naifeng Zhu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Tianhong Tang
- Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhuoli Huang
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Wenbo Jiang
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Junyu Shi
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Yongqiang Hao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Hongchang Lai
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Kerong Dai
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
44
|
Qiao F, Zou Y, Bie B, Lv Y. Dual siRNA-Loaded Cell Membrane Functionalized Matrix Facilitates Bone Regeneration with Angiogenesis and Neurogenesis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307062. [PMID: 37824284 DOI: 10.1002/smll.202307062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/27/2023] [Indexed: 10/14/2023]
Abstract
Vascularization and innervation play irreplaceable roles in bone regeneration and bone defect repair. However, the reconstruction of blood vessels and neural networks is often neglected in material design. This study aims to design a genetically functionalized matrix (GFM) and enable it to regulate angiogenesis and neurogenesis to accelerate the process of bone defect repair. The dual small interfering RNA (siRNA)-polyvinylimide (PEI) (siRP) complexes that locally knocked down soluble vascular endothelial growth factor receptor 1 (sFlt-1) and p75 neurotrophic factor receptor (p75NTR ) are prepared. The hybrid cell membrane (MM) loaded siRP is synthesized as siRNA@MMs to coat on polylactone (PCL) electrospun fibers for mimicking the natural bone matrix. The results indicates that siRNA@MMs could regulate the expression of vascular-related and neuro-related cytokines secreted by mesenchymal stem cells (MSCs). GFMs promote the expression of osteogenic differentiation through paracrine function in vitro. GFMs attenuates inflammation and promotes osseointegration by regulating the coupling of vascularization and innervation in vivo. This study uses the natural hybrid cell membrane to carry genetic material and assist in the vascularization and innervation function of two siRNA. The results present the significance of neuro-vascularized organoid bone and may provide a promising choice for the design of bone tissue engineering scaffold.
Collapse
Affiliation(s)
- Fangyu Qiao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, P. R. China
| | - Yang Zou
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, P. R. China
- School of Environmental Engineering, Wuhan Textile University, Wuhan, 430200, P. R. China
| | - Binglin Bie
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, P. R. China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, P. R. China
| |
Collapse
|
45
|
Zhang Y, Zhu Y, Habibovic P, Wang H. Advanced Synthetic Scaffolds Based on 1D Inorganic Micro-/Nanomaterials for Bone Regeneration. Adv Healthc Mater 2024; 13:e2302664. [PMID: 37902817 DOI: 10.1002/adhm.202302664] [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: 08/13/2023] [Revised: 10/25/2023] [Indexed: 10/31/2023]
Abstract
Inorganic nanoparticulate biomaterials, such as calcium phosphate and bioglass particles, with chemical compositions similar to that of the inorganic component of natural bone, and hence having excellent biocompatibility and bioactivity, are widely used for the fabrication of synthetic bone graft substitutes. Growing evidence suggests that structurally anisotropic, or 1D inorganic micro-/nanobiomaterials are superior to inorganic nanoparticulate biomaterials in the context of mechanical reinforcement and construction of self-supporting 3D network structures. Therefore, in the past decades, efforts have been devoted to developing advanced synthetic scaffolds for bone regeneration using 1D micro-/nanobiomaterials as building blocks. These scaffolds feature extraordinary physical and biological properties, such as enhanced mechanical properties, super elasticity, multiscale hierarchical architecture, extracellular matrix-like fibrous microstructure, and desirable biocompatibility and bioactivity, etc. In this review, an overview of recent progress in the development of advanced scaffolds for bone regeneration is provided based on 1D inorganic micro-/nanobiomaterials with a focus on their structural design, mechanical properties, and bioactivity. The promising perspectives for future research directions are also highlighted.
Collapse
Affiliation(s)
- Yonggang Zhang
- State Key Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China
| | - Yingjie Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Pamela Habibovic
- Maastricht University, Minderbroedersberg 4-6, Maastricht, 6211 LK ER, The Netherlands
| | - Huanan Wang
- State Key Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China
| |
Collapse
|
46
|
Ren Y, Zhang C, Liu Y, Kong W, Yang X, Niu H, Qiang L, Yang H, Yang F, Wang C, Wang J. Advances in 3D Printing of Highly Bioadaptive Bone Tissue Engineering Scaffolds. ACS Biomater Sci Eng 2024; 10:255-270. [PMID: 38118130 DOI: 10.1021/acsbiomaterials.3c01129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The number of patients with bone defects caused by trauma, bone tumors, and osteoporosis has increased considerably. The repair of irregular, recurring, and large bone defects poses a great challenge to clinicians. Bone tissue engineering is emerging as an appropriate strategy to replace autologous bone grafting in the repair of critically sized bone defects. However, the suitability of bone tissue engineering scaffolds in terms of structure, mechanics, degradation, and the microenvironment is inadequate. Three-dimensional (3D) printing is an advanced additive-manufacturing technology widely used for bone repair. 3D printing constructs personalized structurally adapted scaffolds based on 3D models reconstructed from CT images. The contradiction between the mechanics and degradation is resolved by altering the stacking structure. The local microenvironment of the implant is improved by designing an internal pore structure and a spatiotemporal factor release system. Therefore, there has been a boom in the 3D printing of personalized bone repair scaffolds. In this review, successful research on the preparation of highly bioadaptive bone tissue engineering scaffolds using 3D printing is presented. The mechanisms of structural, mechanical, degradation, and microenvironmental adaptations of bone prostheses and their interactions were elucidated to provide a feasible strategy for constructing highly bioadaptive bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Ya Ren
- School of Rehabilitation Medicine, Weifang Medical University, Shandong 261041, China
- Southwest JiaoTong University College of Medicine, No. 111 North first Section of Second Ring Road, Chengdu 610036, China
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Changru Zhang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Weiqing Kong
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266000, Shandong Province, China
| | - Xue Yang
- Southwest JiaoTong University College of Medicine, No. 111 North first Section of Second Ring Road, Chengdu 610036, China
| | - Haoyi Niu
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Lei Qiang
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Han Yang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Fei Yang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Chengwei Wang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Jinwu Wang
- School of Rehabilitation Medicine, Weifang Medical University, Shandong 261041, China
- Southwest JiaoTong University College of Medicine, No. 111 North first Section of Second Ring Road, Chengdu 610036, China
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| |
Collapse
|
47
|
Zhang Y, Ma S, Nie J, Liu Z, Chen F, Li A, Pei D. Journey of Mineral Precursors in Bone Mineralization: Evolution and Inspiration for Biomimetic Design. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2207951. [PMID: 37621037 DOI: 10.1002/smll.202207951] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/27/2023] [Indexed: 08/26/2023]
Abstract
Bone mineralization is a ubiquitous process among vertebrates that involves a dynamic physical/chemical interplay between the organic and inorganic components of bone tissues. It is now well documented that carbonated apatite, an inorganic component of bone, is proceeded through transient amorphous mineral precursors that transforms into the crystalline mineral phase. Here, the evolution on mineral precursors from their sources to the terminus in the bone mineralization process is reviewed. How organisms tightly control each step of mineralization to drive the formation, stabilization, and phase transformation of amorphous mineral precursors in the right place, at the right time, and rate are highlighted. The paradigm shifts in biomineralization and biomaterial design strategies are intertwined, which promotes breakthroughs in biomineralization-inspired material. The design principles and implementation methods of mineral precursor-based biomaterials in bone graft materials such as implant coatings, bone cements, hydrogels, and nanoparticles are detailed in the present manuscript. The biologically controlled mineralization mechanisms will hold promise for overcoming the barriers to the application of biomineralization-inspired biomaterials.
Collapse
Affiliation(s)
- Yuchen Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shaoyang Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiaming Nie
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhongbo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Faming Chen
- School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Dandan Pei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
48
|
Alarçin E, Yaşayan G, Bal-Öztürk A, Cecen B. Hydrogel Biomaterial in Bone Tissue Engineering. BIOMATERIAL-BASED HYDROGELS 2024:387-427. [DOI: 10.1007/978-981-99-8826-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
49
|
Ghosh S, Pati F. Decellularized extracellular matrix and silk fibroin-based hybrid biomaterials: A comprehensive review on fabrication techniques and tissue-specific applications. Int J Biol Macromol 2023; 253:127410. [PMID: 37844823 DOI: 10.1016/j.ijbiomac.2023.127410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Biomaterials play a fundamental role in tissue engineering by providing biochemical and physical cues that influence cellular fate and matrix development. Decellularized extracellular matrix (dECM) as a biomaterial is distinguished by its abundant composition of matrix proteins, such as collagen, elastin, fibronectin, and laminin, as well as glycosaminoglycans and proteoglycans. However, the mechanical properties of only dECM-based constructs may not always meet tissue-specific requirements. Recent advancements address this challenge by utilizing hybrid biomaterials that harness the strengths of silk fibroin (SF), which contributes the necessary mechanical properties, while dECM provides essential cellular cues for in vitro studies and tissue regeneration. This review discusses emerging trends in developing such biopolymer blends, aiming to synergistically combine the advantages of SF and dECM through optimal concentrations and desired cross-linking density. We focus on different fabrication techniques and cross-linking methods that have been utilized to fabricate various tissue-engineered hybrid constructs. Furthermore, we survey recent applications of such biomaterials for the regeneration of various tissues, including bone, cartilage, trachea, bladder, vascular graft, heart, skin, liver, and other soft tissues. Finally, the trajectory and prospects of the constructs derived from this blend in the tissue engineering field have been summarized, highlighting their potential for clinical translation.
Collapse
Affiliation(s)
- Soham Ghosh
- BioFab Lab, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502285, Telangana, India
| | - Falguni Pati
- BioFab Lab, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502285, Telangana, India.
| |
Collapse
|
50
|
Sun W, Ye B, Chen S, Zeng L, Lu H, Wan Y, Gao Q, Chen K, Qu Y, Wu B, Lv X, Guo X. Neuro-bone tissue engineering: emerging mechanisms, potential strategies, and current challenges. Bone Res 2023; 11:65. [PMID: 38123549 PMCID: PMC10733346 DOI: 10.1038/s41413-023-00302-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023] Open
Abstract
The skeleton is a highly innervated organ in which nerve fibers interact with various skeletal cells. Peripheral nerve endings release neurogenic factors and sense skeletal signals, which mediate bone metabolism and skeletal pain. In recent years, bone tissue engineering has increasingly focused on the effects of the nervous system on bone regeneration. Simultaneous regeneration of bone and nerves through the use of materials or by the enhancement of endogenous neurogenic repair signals has been proven to promote functional bone regeneration. Additionally, emerging information on the mechanisms of skeletal interoception and the central nervous system regulation of bone homeostasis provide an opportunity for advancing biomaterials. However, comprehensive reviews of this topic are lacking. Therefore, this review provides an overview of the relationship between nerves and bone regeneration, focusing on tissue engineering applications. We discuss novel regulatory mechanisms and explore innovative approaches based on nerve-bone interactions for bone regeneration. Finally, the challenges and future prospects of this field are briefly discussed.
Collapse
Affiliation(s)
- Wenzhe Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bing Ye
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Siyue Chen
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lian Zeng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hongwei Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yizhou Wan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qing Gao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaifang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanzhen Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bin Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|