1
|
Fang H, Wang Y, Li L, Qin X, Zhu D, Liu P, Yang Q, Gao Y, Shi Z, Ma X, Zhong C, Chen Y. Microenvironment-responsive living hydrogel containing engineered probiotic for treatment of massive bone defects. Bioact Mater 2025; 50:556-570. [PMID: 40385972 PMCID: PMC12083996 DOI: 10.1016/j.bioactmat.2025.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/02/2025] [Accepted: 04/16/2025] [Indexed: 05/20/2025] Open
Abstract
Self-activating and microenvironment-responsive biomaterials for tissue regeneration would address the escalating need for bone grafting, but remain challenging. The emergence of microbial living therapeutics offers vast potential in regenerative medicine, as genetically engineered probiotics possess efficient stimuli-responsiveness and tunable biological functions. Here, using elevated endogenous nitric oxide (NO) signals as a biological trigger in bone fracture injuries, a Living Responsive Regenerative Medicine (LRRM) strategy for in situ bone defect repair through real-time controlled release of bone morphogenetic protein-2 (BMP2) is proposed. The Escherichia coli Nissle 1917 (EcN) strain, genetically engineered to sense NO signals and correspondingly produce and secrete BMP2, was firstly encapsulated in gelatin methacryloyl (GelMA) microspheres and then embedded in a bulky hyaluronic acid methacryloyl (HAMA) hydrogel to form a living hydrogel device that circumvents immune attack and prevents bacterial leakage. In vivo multiple bone defect models demonstrated the efficacy of the living hydrogel in enhancing the maturation of bone callus, promoting neovascularization, and facilitating full-thickness bone union. Strategic incorporation of engineered probiotics and the bilayer-structured encapsulation system may emerge as an effective and microenvironment-responsive medicine approach for tissue regeneration.
Collapse
Affiliation(s)
- Haoyu Fang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yanyi Wang
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
| | - Li Li
- State Key Laboratory of Food Nutrition & Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
- Key Laboratory of Industrial Fermentation Microbiology (Ministry of Education), Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Xiaotong Qin
- State Key Laboratory of Food Nutrition & Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
- Key Laboratory of Industrial Fermentation Microbiology (Ministry of Education), Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Daoyu Zhu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Pei Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qianhao Yang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhongmin Shi
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xin Ma
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chao Zhong
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
| | - Yixuan Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
2
|
Chen S, Wu Z, Huang Z, Liang C, Lee SJ. Implantable Dental Barrier Membranes as Regenerative Medicine in Dentistry: A Comprehensive Review. Tissue Eng Regen Med 2025; 22:527-549. [PMID: 39992621 PMCID: PMC12122982 DOI: 10.1007/s13770-025-00704-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Periodontitis and bone loss in the maxillofacial and dental areas pose considerable challenges for both functional and aesthetic outcomes. To date, implantable dental barrier membranes, designed to prevent epithelial migration into defects and create a favorable environment for targeted cells, have garnered significant interest from researchers. Consequently, a variety of materials and fabrication methods have been explored in extensive research on regenerative dental barrier membranes. METHODS This review focuses on dental barrier membranes, summarizing the various biomaterials used in membrane manufacturing, fabrication methods, and state-of-the-art applications for dental tissue regeneration. Based on a discussion of the pros and cons of current membrane strategies, future research directions for improved membrane designs are proposed. RESULTS AND CONCLUSION To endow dental membranes with various biological properties that accommodate different clinical situations, numerous biomaterials and manufacturing methods have been proposed. These approaches provide theoretical support and hold promise for advancements in dental tissue regeneration.
Collapse
Affiliation(s)
- Siyuan Chen
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, People's Republic of China
| | - Zhenzhen Wu
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, People's Republic of China
| | - Ziqi Huang
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, People's Republic of China
| | - Chao Liang
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, People's Republic of China
| | - Sang Jin Lee
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
3
|
xin D, Junwen Z, Jiacheng Z, Qiongyu X, Huanzong H, peng L, juan L. In vitro and in vivo study of concentrated growth factor (CGF) mediating macrophage polarization in bone defect repair. Regen Ther 2025; 29:474-483. [PMID: 40337617 PMCID: PMC12056952 DOI: 10.1016/j.reth.2025.04.013] [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: 01/08/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
Concentrated growth factor (CGF) is widely applied in clinical practice, but whether it has bone promoting effects and its mechanism of action are still the focus of discussion. In this study, in vitro experiments demonstrate that CGF can promote the expression of Arg-1 in BMDM cells, facilitating their polarization towards the M2 macrophages and encouraging the secretion of IL-10 and VEGF-A. CGF modulates M1 macrophages by reducing the expression of iNOS, while enhancing Arg-1 expression, thereby converting them to M2 macrophages. This is accompanied by a decrease in the secretion of TNF- α and IL-1β, and an increase in the secretion of IL-10 and VEGF-A. Mechanistically, CGF promotes the phosphorylation of STAT3, which in turn induces M2 macrophage polarization, suggesting that the function of CGF-mediated macrophages may be associated with the STAT3 signaling pathway. Moreover, CGF-mediated macrophages were found to enhance osteoblast activity, increasing the expression of ALP, RUNX2, and BMP-2, and improving cell migration capabilities. In vivo experiments showed that CGF could early recruit M2 macrophages to the bone defect site, promoting the expression of bone formation-related proteins such as ALP and BMP-2, and accelerating bone tissue regeneration. In summary, our study demonstrates that CGF can induce bone repair and regeneration by promoting immune modulation and macrophage polarization.
Collapse
Affiliation(s)
- Deng xin
- Department of Stomatology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Zheng Junwen
- Department of Stomatology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Zou Jiacheng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xun Qiongyu
- Pingshan District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - He Huanzong
- Department of Stomatology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Li peng
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Liao juan
- Department of Stomatology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Ma R, Wu Z, Guo X, Wu Z, Zhu Z, Qu Y, Wang K, Li C, Ma K, Yang P. A dual-functional biodegradable composite coating fabricated on sulfonated PEEK via vacuum cold spraying: immunomodulation-driven osteointegration. J Mater Chem B 2025. [PMID: 40423512 DOI: 10.1039/d5tb00628g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Polyetheretherketone (PEEK) is a promising orthopedic implant alternative to metals due to its bone-mimetic modulus and biocompatibility; yet, its bioinert nature often triggers fibrous encapsulation and impedes osteointegration. Here, a biodegradable calcium silicate/β-tricalcium phosphate (CS/TCP) composite coating was fabricated on sulfonated PEEK (SP) via vacuum cold spraying to address these limitations. The CS/TCP coating exhibited robust bonding strength, enhanced hydrophilicity, and sustained release of Ca/Si ions, fostering apatite deposition in simulated body fluid. This bioactive interface promoted an immunomodulatory microenvironment by polarizing macrophages toward the anti-inflammatory M2 phenotype. Synergistically, ionic release and cytokine secretion enhanced MC3T3-E1 cell adhesion, proliferation, and osteogenic differentiation. In vivo, CS/TCP-SP reduced fibrous tissue thickness in a rat air-pouch model and improved bone-implant integration in rabbit cranial defects. The scalable coating strategy transforms inert PEEK into a bioactive, immunoregulatory implant, demonstrating potential to mitigate aseptic loosening and revision surgeries.
Collapse
Affiliation(s)
- Rui Ma
- Joint and Foot & Ankle Ward of Orthopedic Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shanxi, China.
| | - Zidong Wu
- Joint and Foot & Ankle Ward of Orthopedic Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shanxi, China.
| | - Xiaoyu Guo
- Joint and Foot & Ankle Ward of Orthopedic Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shanxi, China.
| | - Zixuan Wu
- Joint and Foot & Ankle Ward of Orthopedic Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shanxi, China.
| | - Zheyue Zhu
- Joint and Foot & Ankle Ward of Orthopedic Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shanxi, China.
| | - Yuning Qu
- Joint and Foot & Ankle Ward of Orthopedic Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shanxi, China.
| | - Kunzheng Wang
- Joint and Foot & Ankle Ward of Orthopedic Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shanxi, China.
| | - Chengxin Li
- State Key Laboratory for Mechanical Behavior of Materials, School of Materials Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Kai Ma
- State Key Laboratory for Mechanical Behavior of Materials, School of Materials Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Pei Yang
- Joint and Foot & Ankle Ward of Orthopedic Center, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shanxi, China.
| |
Collapse
|
5
|
Furko M. Bioglasses Versus Bioactive Calcium Phosphate Derivatives as Advanced Ceramics in Tissue Engineering: Comparative and Comprehensive Study, Current Trends, and Innovative Solutions. J Funct Biomater 2025; 16:161. [PMID: 40422826 DOI: 10.3390/jfb16050161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025] Open
Abstract
Tissue engineering represents a revolutionary approach to regenerating damaged bones and tissues. The most promising materials for this purpose are calcium phosphate-based bioactive ceramics (CaPs) and bioglasses, due to their excellent biocompatibility, osteoconductivity, and bioactivity. This review aims to provide a comprehensive and comparative analysis of different bioactive calcium phosphate derivatives and bioglasses, highlighting their roles and potential in both bone and soft tissue engineering as well as in drug delivery systems. We explore their applications as composites with natural and synthetic biopolymers, which can enhance their mechanical and bioactive properties. This review critically examines the advantages and limitations of each material, their preparation methods, biological efficacy, biodegradability, and practical applications. By summarizing recent research from scientific literature, this paper offers a detailed analysis of the current state of the art. The novelty of this work lies in its systematic comparison of bioactive ceramics and bioglasses, providing insights into their suitability for specific tissue engineering applications. The expected primary outcomes include a deeper understanding of how each material interacts with biological systems, their suitability for specific applications, and the implications for future research directions.
Collapse
Affiliation(s)
- Monika Furko
- Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege Str. 29-33, H-1121 Budapest, Hungary
| |
Collapse
|
6
|
Luo Y, Liu B, Qiu Y, Li L, Yang F, Zhang C, Wang J. Divalent metal ions enhance bone regeneration through modulation of nervous systems and metabolic pathways. Bioact Mater 2025; 47:432-447. [PMID: 40034410 PMCID: PMC11872643 DOI: 10.1016/j.bioactmat.2025.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 03/05/2025] Open
Abstract
The divalent metal cations promote new bone formation through modulation of sensory and sympathetic nervous systems (SNS) activities. In addition, acetylcholine (Ach), as a chief neurotransmitter released by the parasympathetic nervous system (PNS), also affects bone remodeling, so it is of worth to investigate if the divalent cations influence PNS activity. Of note, these cations are key co-enzymes modulating glucose metabolism. Aerobic glycolysis rather than oxidative phosphorylation favors osteogenesis of mesenchymal stem cells (MSCs), so it is of interest to study the effects of these cations on glucose metabolic pathway. Prior to biological function assessment, the tolerance limits of the divalent metal cations (Mg2+, Zn2+, and Ca2+) and their combinations were profiled. In terms of direct effects, these divalent cations potentially enhanced migration and adhesion capability of MSCs through upregulating Tgf-β1 and Integrin-β1 levels. Interestingly, the divalent cations alone did not influence osteogenesis and aerobic glycolysis of undifferentiated MSCs. However, once the osteogenic differentiation of MSCs was initiated by neurotransmitters or osteogenic differentiation medium, the osteogenesis of MSCs could be significantly promoted by the divalent cations, which was accompanied by the improved aerobic glycolysis. In terms of indirect effects, the divalent cations significantly upregulated levels of sensory nerve derived CGRP, PNS produced choline acetyltransferase and type H vessels, while significantly tuned down sympathetic activity in the defect zone in rats, thereby contributing to significantly increased bone formation relative to the control group. Together, the divalent cations favor bone regeneration via modulation of sensory-autonomic nervous systems and promotion of aerobic glycolysis-driven osteogenesis of MSCs after osteogenic initiation by neurotransmitters.
Collapse
Affiliation(s)
- Ying Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Baoyi Liu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, Liaoning, China
| | - Yashi Qiu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Lichen Li
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Fan Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, Liaoning, China
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Jiali Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
7
|
Jo S, Hwangbo H, Francis N, Lee J, Pei M, Kim G. Fish-derived biomaterials for tissue engineering: advances in scaffold fabrication and applications in regenerative medicine and cancer therapy. Theranostics 2025; 15:5666-5692. [PMID: 40365274 PMCID: PMC12068294 DOI: 10.7150/thno.109186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/10/2025] [Indexed: 05/15/2025] Open
Abstract
Fish-derived biomaterials, such as collagen, polyunsaturated fatty acids, and antimicrobial peptides, have emerged as promising candidates for scaffold development in stem cell therapies and tissue engineering due to their excellent biocompatibility and low immunogenicity. Although good bioactivity is a prerequisite for biomedical substitutes, scaffold design is necessary for the successful development of bioconstructs used in tissue regeneration. However, the limited processability of fish biomaterials poses a substantial challenge to the development of diverse scaffold structures. In this review, unlike previous reviews that primarily focused on the bioactivities of fish-derived components, we placed greater emphasis on scaffold fabrication and its applications in tissue regeneration. Specifically, we examined various cross-linking strategies to enhance the structural integrity of fish biomaterials and address challenges, such as poor processability, low mechanical strength, and rapid degradation. Furthermore, we demonstrated the potential of fish scaffolds in stem cell therapies, particularly their capacity to support stem cell growth and modulate the cellular microenvironment. Finally, this review provides future directions for the application of these scaffolds in cancer therapy.
Collapse
Affiliation(s)
- Seoyul Jo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - Hanjun Hwangbo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - Nacionales Francis
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - JaeYoon Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - Mohan Pei
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - GeunHyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
8
|
Kabir A, Mukilarasi B, Manohar A, Gadani M, Sinha AK, Sharma P, Verma A, Selvaraj V, Sudhakar S. Protein bioactive complexes promote osteogenesis under microgravity environment. Int J Biol Macromol 2025; 303:140483. [PMID: 39904451 DOI: 10.1016/j.ijbiomac.2025.140483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025]
Abstract
The space microgravity environment and cosmic radiation pose a significant threat to musculoskeletal health, particularly bone mass. However, the critical mechanism underlying space-induced bone loss and its relation to cellular oxidative stress remains unclear. Currently used bone-loss-reversing drugs face limitations like poor efficacy and metabolic defects. Herein, we revealed that simulated microgravity (SMG) induces reactive oxygen species (ROS), negatively impacting osteoblasts, causing cytoskeletal damage, and downregulating osteogenic genes. To combat this, we designed protein-zein nanocages loaded with a chimeric non-enzymatic cocktail (ZNAC) containing ascorbic acid, resveratrol, luteolin, coenzyme Q, and glutathione. These nanocages (~200 nm) demonstrated excellent stability, biocompatibility, and antioxidant properties compared to free drugs. We investigated the effects of ZNAC under SMG using two experimental models: MC3T3 pre-osteoblast/MG63 osteoblasts and regenerating zebrafish scales that represent compositional and physiological/pathophysiological analogy with mammalian system. ZNAC effectively reduced SMG-induced ROS, preserved cytoskeletal integrity, and enhanced alkaline phosphatase (ALP) activity along with the expression of osteogenic genes such as RUNX2 and Col1A1. In zebrafish scales, it increased osteogenic gene expression, calcification, and the calcium/phosphorus ratio, indicating enhanced scale regeneration. These findings suggest that ZNAC is a promising candidate for bone regeneration, offering potential solutions for maintaining astronaut health during extended space missions.
Collapse
Affiliation(s)
- Anisha Kabir
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - B Mukilarasi
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - Anagha Manohar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - Maulesh Gadani
- Space Applications Centre, Indian Space Research Organisation, Ahmedabad, Gujarat 380015, India.
| | - Anurag Kumar Sinha
- Human Space Flight Centre, Antariksh Bhavan, New BEL Road, Bengaluru 560 094, India.
| | - Payal Sharma
- Space Applications Centre, Indian Space Research Organisation, Ahmedabad, Gujarat 380015, India.
| | - Anurag Verma
- Space Applications Centre, Indian Space Research Organisation, Ahmedabad, Gujarat 380015, India.
| | - Vimalraj Selvaraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - Swathi Sudhakar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| |
Collapse
|
9
|
Choudhury S, Das D, Roy S, Chowdhury AR. Piezoelectric Biomaterials for Use in Bone Tissue Engineering-A Narrative Review. J Biomed Mater Res B Appl Biomater 2025; 113:e35564. [PMID: 40096659 DOI: 10.1002/jbm.b.35564] [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: 10/26/2024] [Revised: 02/03/2025] [Accepted: 02/28/2025] [Indexed: 03/19/2025]
Abstract
To examine natural bone's bioelectrical traits, notably its piezoelectricity, and to look into how these characteristics influence bone growth and repair. In the context of exploring the potential of piezoelectric biomaterials, such as biopolymers and bio-ceramics, towards orthopedic and bone regeneration applications, the research seeks to evaluate the significance of piezoelectricity-driven osteogenesis. The paper reviews recent research on bone's electrical and dielectric properties, surface polarization/electrical stimulation effects interacting with cell activity and the effectiveness of piezoelectric biomaterials to support tissues' regenerative process. The study includes a number of materials, such as collagen, polyvinylidene fluoride (PVDF) and barium titanate. The applications of piezoelectric bio-ceramics, piezoelectric organic polymers, and piezoelectric natural polymers are particularly highlighted. Piezoelectric biomaterials are being shown in recent studies to enhance cellular metabolism in vitro as well as promote the regeneration of tissues in vivo, especially when paired with electric field stimulation or interface polarization. Piezoelectric bio-ceramics like magnesium silicate and barium titanate, as well as biopolymers like collagen and PVDF, have shown possibilities for orthopedic applications. However, there are several challenges regarding the manufacturing of bio-ceramics of specific compositions having the desired properties. This review highlighted the potential of piezoelectric biomaterials in orthopedic applications with special emphasis on biopolymers and bioceramics. Therefore, these types of materials have huge potential for bone regeneration because they can mimic the piezoelectric properties of bone and allow better advances in tissue engineering or regenerative medicine. To date, little is known about their mechanism of action, and modifications are needed to improve efficacy for clinical uptake.
Collapse
Affiliation(s)
- Sandeep Choudhury
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Howrah, West Bengal, India
| | - Debolina Das
- Department of Mechanical Engineering, Indian Institute of Engineering Science and Technology, Howrah, West Bengal, India
| | - Sandipan Roy
- Department of Mechanical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Amit Roy Chowdhury
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Howrah, West Bengal, India
| |
Collapse
|
10
|
Rizzo MG, Briglia M, Zammuto V, Morganti D, Faggio C, Impellitteri F, Multisanti CR, Graziano ACE. Innovation in Osteogenesis Activation: Role of Marine-Derived Materials in Bone Regeneration. Curr Issues Mol Biol 2025; 47:175. [PMID: 40136429 PMCID: PMC11941683 DOI: 10.3390/cimb47030175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Marine-derived biomaterials are emerging as promising candidates for tissue regeneration due to their sustainability, biocompatibility, bioactivity, and unique chemical structure. This review provides an overview of different marine-derived inorganic and organic materials, such as calcium carbonate, magnesium salts, silica, polysaccharides, bioactive peptides, and lipid-based compounds, and their effects in promoting osteogenesis. Specifically, the osteoinductive, osteoconductive, and osteointegrative activities of traditional and innovative materials that influence key molecular pathways such as BMP/Smad and Wnt/β-catenin signaling underlying bone formation will be evaluated. This review also prospects innovative approaches, i.e., phage display technology, to optimize marine-derived peptides for targeted bone regeneration. In the context of innovative and sustainable materials, this review suggests some interesting applications of unusual materials able to overcome the limitations of conventional ones and stimulate cellular regeneration of bone tissue by activating specific molecular pathways.
Collapse
Affiliation(s)
- Maria Giovanna Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy;
| | - Marilena Briglia
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (A.C.E.G.)
| | - Vincenzo Zammuto
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy;
| | - Dario Morganti
- Consiglio Nazionale delle Ricerche DSFTM, Department of Physical Sciences and Technologies of Matter, Piazzale Aldo Moro, 7, 00185 Roma, Italy
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy;
- Department of Eco-Sustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, 80122 Naples, Italy
| | - Federica Impellitteri
- Department of Veterinary Sciences, University of Messina, Viale Giovanni Palatucci, 98168 Messina, Italy; (F.I.); (C.R.M.)
| | - Cristiana Roberta Multisanti
- Department of Veterinary Sciences, University of Messina, Viale Giovanni Palatucci, 98168 Messina, Italy; (F.I.); (C.R.M.)
| | | |
Collapse
|
11
|
Qin Y, Jing Z, Zou D, Wang Y, Yang H, Chen K, Li W, Wen P, Zheng Y. A metamaterial scaffold beyond modulus limits: enhanced osteogenesis and angiogenesis of critical bone defects. Nat Commun 2025; 16:2180. [PMID: 40038291 PMCID: PMC11880532 DOI: 10.1038/s41467-025-57609-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 02/20/2025] [Indexed: 03/06/2025] Open
Abstract
Metallic scaffolds have shown promise in regenerating critical bone defects. However, limitations persist in achieving a modulus below 100 MPa due to insufficient strength. Consequently, the osteogenic impact of lower modulus and greater bone tissue strain ( > 1%) remains unclear. Here, we introduce a metamaterial scaffold that decouples strength and modulus through two-stage deformation. The scaffold facilitates an effective modulus of only 13 MPa, ensuring adaptability during bone regeneration. Followed by a stiff stage, it provides the necessary strength for load-bearing requirements. In vivo, the scaffold induces > 2% callus strain, upregulating calcium channels and HIF-1α to enhance osteogenesis and angiogenesis. 4-week histomorphology reveals a 44% and 498% increase in new bone fraction versus classic scaffolds with 500 MPa and 13 MPa modulus, respectively. This design transcends traditional modulus-matching paradigms, prioritizing bone tissue strain requirements. Its tunable mechanical properties also present promising implications for advancing osteogenesis mechanisms and addressing clinical challenges.
Collapse
Affiliation(s)
- Yu Qin
- School of Materials Science and Engineering, Peking University, Beijing, China
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Zehao Jing
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Da Zou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Youhao Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Hongtao Yang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Kai Chen
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Weishi Li
- Department of Orthopedics, Peking University Third Hospital, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China.
| | - Peng Wen
- Department of Mechanical Engineering, Tsinghua University, Beijing, China.
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, China.
| |
Collapse
|
12
|
Wen Z, Wu F, Shi J, Cheng H, Xie S, Liang D, Li J, Lu Y. CircFak promotes mechanical force-induced osteogenesis via FAK/AKT phosphorylation. J Dent 2025; 154:105602. [PMID: 39894158 DOI: 10.1016/j.jdent.2025.105602] [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: 11/03/2024] [Revised: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025] Open
Abstract
OBJECTIVES Orthodontic treatment is widely applied for addressing orofacial skeletal deformities, with the remodeling of the alveolar bone under mechanical force being the key factor. FAK is essential for cellular response to mechanical force. However, the function of circFak has never been reported. In this study, the microarrays showed that circFak may affect osteogenesis under mechanical force. We aimed to verify the effect of circFak in force-related bone remodeling and investigate the underlying mechanisms. METHODS Arraystar microarrays were used to identify differentially expressed circRNAs and microRNAs in response to mechanical stress. The subcellular distribution of circFak was analyzed via RT‒qPCR and FISH. ALP and ARS staining assays were performed to investigate the effects of circFak on osteogenesis. RNA sequencing, bioinformatics analysis, dual-luciferase reporter assays, and RNA immunoprecipitation were accomplished to discover the molecular mechanisms of circFak. AAV-sh-circFak mouse models with tooth movements were established. The role of circFak under mechanical force in vivo was assessed via immunofluorescence and micro-CT analyses. RESULTS CircFak expression was significantly upregulated under mechanical force. Osteogenic capacity of osteoblasts was positively correlated with the level of circFak. CircFak promoted mechanical force-induced osteogenesis through miR-425-5p/Ccn3 pathway, and further stimulated the phosphorylation of its parental sourced protein FAK. Our murine models showed that AAV-mediated circFak silencing suppressed osteogenesis. CONCLUSION CircFak could obviously promote osteogenesis under mechanical force and may possess ability to become a novel biomarker for prognosis of orthodontic treatments.
Collapse
Affiliation(s)
- Zhihui Wen
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Fan Wu
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Juanyi Shi
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China; Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huilin Cheng
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Shule Xie
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Defeng Liang
- Department of Stomatology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Jinsong Li
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China.
| | - Yingjuan Lu
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
13
|
Wang L, Ruan M, Bu Q, Zhao C. Signaling Pathways Driving MSC Osteogenesis: Mechanisms, Regulation, and Translational Applications. Int J Mol Sci 2025; 26:1311. [PMID: 39941080 PMCID: PMC11818554 DOI: 10.3390/ijms26031311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are crucial for skeletal development, homeostasis, and repair, primarily through their differentiation into osteoblasts and other skeletal lineage cells. Key signaling pathways, including Wnt, TGF-β/BMP, PTH, Hedgehog, and IGF, act as critical regulators of MSC osteogenesis, playing pivotal roles in maintaining bone homeostasis and facilitating regeneration. These pathways interact in distinct ways at various stages of bone development, mineralization, and remodeling. This review provides an overview of the molecular mechanisms by which these pathways regulate MSC osteogenesis, their influence on bone tissue formation, and their implications in bone diseases and therapeutic strategies. Additionally, we explore the potential applications of these pathways in bone tissue engineering, with a particular focus on promoting the use of MSCs as seed cells for bone defect repair. Ultimately, this review aims to highlight potential avenues for advancing bone biology research, treating bone disorders, and enhancing regenerative medicine.
Collapse
Affiliation(s)
| | | | | | - Chengzhu Zhao
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
14
|
Zhao Z, Feng X, Zhao Y, Song Z, Zhang R, Zhang K, He Y, Chen G, Zhang J, Wang W. Gelatin/Poly (Lactic-Co-Glycolic Acid)/Attapulgite Composite Scaffold Equipped with Teriparatide Microspheres for Osteogenesis in vitro and in vivo. Int J Nanomedicine 2025; 20:581-604. [PMID: 39839456 PMCID: PMC11747967 DOI: 10.2147/ijn.s495204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
Background Given the risks associated with autologous bone transplantation and the limitations of allogeneic bone transplantation, scaffolds in bone tissue engineering that incorporate bioactive peptides are highly recommended. Teriparatide (TPTD) plays a significant role in bone defect repair, although achieving controlled release of TPTD within a bone tissue engineering scaffold remains challenging. This work reports a new approach for treatment of teriparatide using a water-in-oil-in-water (w/o/w) microspheres be equipped on gelatin (GEL)/Poly lactic-glycolic acid (PLGA)/attapulgite (ATP) scaffold. Methods In this study, TPTD microspheres were prepared by the water-in-oil-in-water (w/o/w) double emulsion technique and GEL/PLGA/ATP composite scaffolds with different setups were prepared by salt leaching method. Both microspheres and scaffolds underwent physicochemical characterization. Mouse bone mesenchymal stem cells (BMSCs) were co-cultured with extracts from the microspheres and scaffolds to evaluate cell proliferation and osteogenesis. Four weeks post-implantation, the effectiveness of the scaffolds containing microspheres for repairing skull defects in mice was assessed. Results Both TPTD microspheres and the GEL/PLGA/ATP scaffold significantly enhanced the proliferation and osteogenic differentiation of BMSCs. Markers of osteoblast activity, including COL1, RUNX2, OCN, and OPN, were markedly up-regulated. Further, micro-CT, histological, and immunohistochemical analyses revealed extensive new bone formation on the scaffold. Conclusion The GEL/PLGA/ATP composite scaffold, equipped with TPTD microspheres, demonstrates significant potential for use in bone tissue engineering, providing an effective option for bone regeneration and repair in clinical applications.
Collapse
Affiliation(s)
- Zhenrui Zhao
- Department of Orthopedics, The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Xiaofei Feng
- Department of Orthopedics, The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Yuhao Zhao
- Department of Orthopedics, The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Zhengdong Song
- Department of Orthopedics, The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Ruihao Zhang
- Department of Orthopedics, The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Kui Zhang
- Department of Orthopedics, The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Yixiang He
- Department of Orthopedics, The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Guoliang Chen
- Department of Orthopedics, The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Jing Zhang
- Department of Orthopedics, Anlu People’s Hospital, Anlu, People’s Republic of China
| | - Wenji Wang
- Department of Orthopedics, the First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| |
Collapse
|
15
|
Shariati K, Bedar M, Huang KX, Moghadam S, Mirzaie S, LaGuardia JS, Chen W, Kang Y, Ren X, Lee JC. Biomaterial Cues for Regulation of Osteoclast Differentiation and Function in Bone Regeneration. ADVANCED THERAPEUTICS 2025; 8:2400296. [PMID: 39867107 PMCID: PMC11756815 DOI: 10.1002/adtp.202400296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Indexed: 01/28/2025]
Abstract
Tissue regeneration involves dynamic dialogue between and among different cells and their surrounding matrices. Bone regeneration is specifically governed by reciprocity between osteoblasts and osteoclasts within the bone microenvironment. Osteoclast-directed resorption and osteoblast-directed formation of bone are essential to bone remodeling, and the crosstalk between these cells is vital to curating a sequence of events that culminate in the creation of bone tissue. Among bone biomaterial strategies, many have investigated the use of different material cues to direct the development and activity of osteoblasts. However, less attention has been given to exploring features that similarly target osteoclast formation and activity, with even fewer strategies demonstrating or integrating biomaterial-directed modulation of osteoblast-osteoclast coupling. This review aims to describe various biomaterial cues demonstrated to influence osteoclastogenesis and osteoclast function, emphasizing those that enhance a material construct's ability to achieve bone healing and regeneration. Additionally discussed are approaches that influence the communication between osteoclasts and osteoblasts, particularly in a manner that takes advantage of their coupling. Deepening our understanding of how biomaterial cues may dictate osteoclast differentiation, function, and influence on the microenvironment may enable the realization of bone-replacement interventions with enhanced integrative and regenerative capacities.
Collapse
Affiliation(s)
- Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Sarah Mirzaie
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
16
|
Zheng R, Yu C, Yao D, Cai M, Zhang L, Ye F, Huang X. Engineering Stimuli-Responsive Materials for Precision Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406439. [PMID: 39444066 PMCID: PMC11707583 DOI: 10.1002/smll.202406439] [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: 07/28/2024] [Revised: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Over the past decade, precision medicine has garnered increasing attention, making significant strides in discovering new therapeutic drugs and mechanisms, resulting in notable achievements in symptom alleviation, pain reduction, and extended survival rates. However, the limited target specificity of primary drugs and inter-individual differences have often necessitated high-dosage strategies, leading to challenges such as restricted deep tissue penetration rates and systemic side effects. Material science advancements present a promising avenue for these issues. By leveraging the distinct internal features of diseased regions and the application of specific external stimuli, responsive materials can be tailored to achieve targeted delivery, controllable release, and specific biochemical reactions. This review aims to highlight the latest advancements in stimuli-responsive materials and their potential in precision medicine. Initially, we introduce disease-related internal stimuli and capable external stimuli, elucidating the reaction principles of responsive functional groups. Subsequently, we provide a detailed analysis of representative pre-clinical achievements of stimuli responsive materials across various clinical applications, including enhancements in the treatment of cancers, injury diseases, inflammatory diseases, infection diseases, and high-throughput microfluidic biosensors. Finally, we discuss some clinical challenges, such as off-target effects, long-term impacts of nano-materials, potential ethical concerns, and offer insights into future perspectives of stimuli-responsive materials.
Collapse
Affiliation(s)
- Ruixuan Zheng
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Chang Yu
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
- Intervention DepartmentThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Dan Yao
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Mengsi Cai
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Lexiang Zhang
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000China
| | - Fangfu Ye
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000China
- Beijing National Laboratory for Condensed Matter PhysicsInstitute of PhysicsChinese Academy of SciencesBeijing100190China
| | - Xiaoying Huang
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| |
Collapse
|
17
|
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
|
18
|
Li M, Wu H, Gao K, Wang Y, Hu J, Guo Z, Hu R, Zhang M, Pang X, Guo M, Liu Y, Zhao L, He W, Ding S, Li W, Cheng W. Smart Implantable Hydrogel for Large Segmental Bone Regeneration. Adv Healthc Mater 2024; 13:e2402916. [PMID: 39344873 DOI: 10.1002/adhm.202402916] [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: 08/06/2024] [Revised: 09/16/2024] [Indexed: 10/01/2024]
Abstract
Large segmental bone defects often lead to nonunion and dysfunction, posing a significant challenge for clinicians. Inspired by the intrinsic bone defect repair logic of "vascularization and then osteogenesis", this study originally reports a smart implantable hydrogel (PDS-DC) with high mechanical properties, controllable scaffold degradation, and timing drug release that can proactively match different bone healing cycles to efficiently promote bone regeneration. The main scaffold of PDS-DC consists of polyacrylamide, polydopamine, and silk fibroin, which endows it with superior interfacial adhesion, structural toughness, and mechanical stiffness. In particular, the adjustment of scaffold cross-linking agent mixing ratio can effectively regulate the in vivo degradation rate of PDS-DC and intelligently satisfy the requirements of different bone defect healing cycles. Ultimately, PDS hydrogel loaded with free desferrioxamine (DFO) and CaCO3 mineralized ZIF-90 loaded bone morphogenetic protein-2 (BMP-2) to stimulate efficient angiogenesis and osteogenesis. Notably, DFO is released rapidly by free diffusion, whereas BMP-2 is released slowly by pH-dependent layer-by-layer disintegration, resulting in a significant difference in release time, thus matching the intrinsic logic of bone defect repair. In vivo and in vitro results confirm that PDS-DC can effectively realize high-quality bone generation and intelligently regulate to adapt to different demands of bone defects.
Collapse
Affiliation(s)
- Menghan Li
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Haiping Wu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, P. R. China
| | - Ke Gao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yubo Wang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Jiaqi Hu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Ziling Guo
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing Key Laboratory of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Ruiwei Hu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Mengxuan Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Xiaoxiao Pang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Minghui Guo
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yuanjie Liu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Lina Zhao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Wen He
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Wenyang Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Wei Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| |
Collapse
|
19
|
Donati L, Valicenti ML, Giannoni S, Morena F, Martino S. Biomaterials Mimicking Mechanobiology: A Specific Design for a Specific Biological Application. Int J Mol Sci 2024; 25:10386. [PMID: 39408716 PMCID: PMC11476540 DOI: 10.3390/ijms251910386] [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: 08/10/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Mechanosensing and mechanotransduction pathways between the Extracellular Matrix (ECM) and cells form the essential crosstalk that regulates cell homeostasis, tissue development, morphology, maintenance, and function. Understanding these mechanisms involves creating an appropriate cell support that elicits signals to guide cellular functions. In this context, polymers can serve as ideal molecules for producing biomaterials designed to mimic the characteristics of the ECM, thereby triggering responsive mechanisms that closely resemble those induced by a natural physiological system. The generated specific stimuli depend on the different natural or synthetic origins of the polymers, the chemical composition, the assembly structure, and the physical and surface properties of biomaterials. This review discusses the most widely used polymers and their customization to develop biomaterials with tailored properties. It examines how the characteristics of biomaterials-based polymers can be harnessed to replicate the functions of biological cells, making them suitable for biomedical and biotechnological applications.
Collapse
Affiliation(s)
- Leonardo Donati
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Maria Luisa Valicenti
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Samuele Giannoni
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
- Centro di Eccellenza Materiali Innovativi Nanostrutturati per Applicazioni Chimiche Fisiche e Biomediche (CEMIN), University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
20
|
Chen W, Sheng S, Tan K, Wang S, Wu X, Yang J, Hu Y, Cao L, Xu K, Zhou F, Su J, Zhang Q, Yang L. Injectable hydrogels for bone regeneration with tunable degradability via peptide chirality modification. MATERIALS HORIZONS 2024; 11:4367-4377. [PMID: 38932613 DOI: 10.1039/d4mh00398e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
The degradability of hydrogels plays a pivotal role in bone regeneration, yet its precise effects on the bone repair process remain poorly understood. Traditional studies have been limited by the use of hydrogels with insufficient variation in degradation properties for thorough comparative analysis. Addressing this gap, our study introduces the development of matrix metalloproteinase (MMP)-responsive hydrogels engineered with a tunable degradation rate, specifically designed for bone regeneration applications. These innovative hydrogels are synthesized by integrating MMP-sensitive peptides, which exhibit chirality-transferred amino acids, with norbornene (NB)-modified 8-arm polyethylene glycol (PEG) macromers to form the hydrogel network. The degradation behavior of these hydrogels is manipulated through the chirality of the incorporated peptides, resulting in the classification into L, LD, and D hydrogels. Remarkably, the L hydrogel variant shows a significantly enhanced degradation rate, both in vitro and in vivo, which in turn fosters bone regeneration by promoting cell migration and upregulating osteogenic gene expression. This research highlights the fundamental role of hydrogel degradability in bone repair and lays the groundwork for the advancement of degradable hydrogel technologies for bone regeneration, offering new insights and potential for future biomaterials development.
Collapse
Affiliation(s)
- Weikai Chen
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P. R. China.
- Organoid Research Center, Shanghai University, Shanghai 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P. R. China
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325003, P. R. China.
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang 325035, P. R. China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P. R. China
| | - Kai Tan
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325003, P. R. China.
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang 325035, P. R. China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P. R. China.
- Organoid Research Center, Shanghai University, Shanghai 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P. R. China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, P. R. China
| | - Xiang Wu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P. R. China.
- Organoid Research Center, Shanghai University, Shanghai 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P. R. China
| | - Jiayi Yang
- Department of Gynaecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325003, P. R. China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P. R. China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Luodian Hospital, Shanghai 201908, P. R. China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P. R. China.
- Organoid Research Center, Shanghai University, Shanghai 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P. R. China
| | - Fengjin Zhou
- Honghui Hospital, Xi'an Jiao Tong University, Xi'an 710000, P. R. China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P. R. China.
- Organoid Research Center, Shanghai University, Shanghai 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P. R. China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P. R. China
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P. R. China.
- Organoid Research Center, Shanghai University, Shanghai 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P. R. China
| | - Lei Yang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325003, P. R. China.
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang 325035, P. R. China
| |
Collapse
|
21
|
Pan P, Wang J, Wang X, Yu X, Chen T, Jiang C, Liu W. Barrier Membrane with Janus Function and Structure for Guided Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47178-47191. [PMID: 39222394 DOI: 10.1021/acsami.4c08737] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Guided bone regeneration (GBR) technology has been demonstrated to be an effective method for reconstructing bone defects. A membrane is used to cover the bone defect to stop soft tissue from growing into it. The biosurface design of the barrier membrane is key to the technology. In this work, an asymmetric functional gradient Janus membrane was designed to address the bidirectional environment of the bone and soft tissue during bone reconstruction. The Janus membrane was simply and efficiently prepared by the multilayer self-assembly technique, and it was divided into the polycaprolactone isolation layer (PCL layer, GBR-A) and the nanohydroxyapatite/polycaprolactone/polyethylene glycol osteogenic layer (HAn/PCL/PEG layer, GBR-B). The morphology, composition, roughness, hydrophilicity, biocompatibility, cell attachment, and osteogenic mineralization ability of the double surfaces of the Janus membrane were systematically evaluated. The GBR-A layer was smooth, dense, and hydrophobic, which could inhibit cell adhesion and resist soft tissue invasion. The GBR-B layer was rough, porous, hydrophilic, and bioactive, promoting cell adhesion, proliferation, matrix mineralization, and expression of alkaline phosphatase and RUNX2. In vitro and in vivo results showed that the membrane could bind tightly to bone, maintain long-term space stability, and significantly promote new bone formation. Moreover, the membrane could fix the bone filling material in the defect for a better healing effect. This work presents a straightforward and viable methodology for the fabrication of GBR membranes with Janus-based bioactive surfaces. This work may provide insights for the design of biomaterial surfaces and treatment of bone defects.
Collapse
Affiliation(s)
- Peng Pan
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, P. R. China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Jian Wang
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Xi Wang
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, P. R. China
| | - Xinding Yu
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, P. R. China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Tiantian Chen
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, P. R. China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Chundong Jiang
- Chongqing Institute of Mesoscopic Medical Porous Materials, Chongqing 401120, P. R. China
| | - Wentao Liu
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, P. R. China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| |
Collapse
|
22
|
Wang Y, Lv H, Ren S, Zhang J, Liu X, Chen S, Zhai J, Zhou Y. Biological Functions of Macromolecular Protein Hydrogels in Constructing Osteogenic Microenvironment. ACS Biomater Sci Eng 2024; 10:5513-5536. [PMID: 39173130 DOI: 10.1021/acsbiomaterials.4c00910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Irreversible bone defects resulting from trauma, infection, and degenerative illnesses have emerged as a significant health concern. Structurally and functionally controllable hydrogels made by bone tissue engineering (BTE) have become promising biomaterials. Natural proteins are able to establish connections with autologous proteins through unique biologically active regions. Hydrogels based on proteins can simulate the bone microenvironment and regulate the biological behavior of stem cells in the tissue niche, making them candidates for research related to bone regeneration. This article reviews the biological functions of various natural macromolecular proteins (such as collagen, gelatin, fibrin, and silk fibroin) and highlights their special advantages as hydrogels. Then the latest research trends on cross-linking modified macromolecular protein hydrogels with improved mechanical properties and composite hydrogels loaded with exogenous micromolecular proteins have been discussed. Finally, the applications of protein hydrogels, such as 3D printed hydrogels, microspheres, and injectable hydrogels, were introduced, aiming to provide a reference for the repair of clinical bone defects.
Collapse
Affiliation(s)
- Yihan Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Huixin Lv
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Sicong Ren
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Jiameng Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Xiuyu Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Sheng Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Jingjie Zhai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| |
Collapse
|
23
|
Liu Y, Li L, He M, Xu Y, Wu Z, Xu X, Luo K, Lv H. Self-assembled peptide hydrogel loaded with functional peptide Dentonin accelerates vascularized bone tissue regeneration in critical-size bone defects. Regen Biomater 2024; 11:rbae106. [PMID: 39263324 PMCID: PMC11387769 DOI: 10.1093/rb/rbae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/18/2024] [Accepted: 08/18/2024] [Indexed: 09/13/2024] Open
Abstract
Regeneration of oral craniofacial bone defects is a complex process, and reconstruction of large bone defects without the use of exogenous cells or bioactive substances remains a major challenge. Hydrogels are highly hydrophilic polymer networks with the potential to promote bone tissue regeneration. In this study, functional peptide Dentonin was loaded onto self-assembled peptide hydrogels (RAD) to constitute functionally self-assembling peptide RAD/Dentonin hydrogel scaffolds with a view that RAD/Dentonin hydrogel could facilitate vascularized bone regeneration in critical-size calvarial defects. The functionalized peptide RAD/Dentonin forms highly ordered β-sheet supramolecular structures via non-covalent interactions like hydrogen bonding, ultimately assembling into nano-fiber network. RAD/Dentonin hydrogels exhibited desirable porosity and swelling properties, and appropriate biodegradability. RAD/Dentonin hydrogel supported the adhesion, proliferation and three-dimensional migration of bone marrow mesenchymal stem cells (BMSCs) and has the potential to induce differentiation of BMSCs towards osteogenesis through activation of the Wnt/β-catenin pathway. Moreover, RAD/Dentonin hydrogel modulated paracrine secretion of BMSCs and increased the migration, tube formation and angiogenic gene expression of human umbilical vein endothelial cells (HUVECs), which boosted the angiogenic capacity of HUVECs. In vivo, RAD/Dentonin hydrogel significantly strengthened vascularized bone formation in rat calvarial defect. Taken together, these results indicated that the functionalized self-assembling peptide RAD/Dentonin hydrogel effectively enhance osteogenic differentiation of BMSCs, indirectly induce angiogenic effects in HUVECs, and facilitate vascularized bone regeneration in vivo. Thus, it is a promising bioactive material for oral and maxillofacial regeneration.
Collapse
Affiliation(s)
- Yijuan Liu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Li Li
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Mengjiao He
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Yanmei Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Zekai Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Hongbing Lv
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| |
Collapse
|
24
|
Jiang K, Wang K, Luo C, Su BY, Du H, Liu Y, Lei J, Luo E, Cardon L, Edeleva M, Huang SS, Xu JZ, Li ZM. A Biomimetic Fibrous Composite Scaffold with Nanotopography-Regulated Mineralization for Bone Defect Repair. Biomacromolecules 2024; 25:3784-3794. [PMID: 38743836 DOI: 10.1021/acs.biomac.4c00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The effective regeneration of large bone defects via bone tissue engineering is challenging due to the difficulty in creating an osteogenic microenvironment. Inspired by the fibrillar architecture of the natural extracellular matrix, we developed a nanoscale bioengineering strategy to produce bone fibril-like composite scaffolds with enhanced osteogenic capability. To activate the surface for biofunctionalization, self-adaptive ridge-like nanolamellae were constructed on poly(ε-caprolactone) (PCL) electrospinning scaffolds via surface-directed epitaxial crystallization. This unique nanotopography with a markedly increased specific surface area offered abundant nucleation sites for Ca2+ recruitment, leading to a 5-fold greater deposition weight of hydroxyapatite than that of the pristine PCL scaffold under stimulated physiological conditions. Bone marrow mesenchymal stem cells (BMSCs) cultured on bone fibril-like scaffolds exhibited enhanced adhesion, proliferation, and osteogenic differentiation in vitro. In a rat calvarial defect model, the bone fibril-like scaffold significantly accelerated bone regeneration, as evidenced by micro-CT, histological histological and immunofluorescence staining. This work provides the way for recapitulating the osteogenic microenvironment in tissue-engineered scaffolds for bone repair.
Collapse
Affiliation(s)
- Kai Jiang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Kai Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Chuan Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Biao-Yao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hao Du
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - Jun Lei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - Ludwig Cardon
- Centre for Polymer and Material Technologies, Department of Materials Textiles and Chemical Engineering, Ghent University, Technologiepark-Zwijnaarde 130, Gent 9052, Belgium
| | - Mariya Edeleva
- Centre for Polymer and Material Technologies, Department of Materials Textiles and Chemical Engineering, Ghent University, Technologiepark-Zwijnaarde 130, Gent 9052, Belgium
| | - Shi-Shu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhong-Ming Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
25
|
Vermeulen S, Balmayor ER. Discovering the nucleus in a world of biomaterials. BIOMATERIALS AND BIOSYSTEMS 2024; 14:100096. [PMID: 38974419 PMCID: PMC11225202 DOI: 10.1016/j.bbiosy.2024.100096] [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: 01/08/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
The nucleus serves as the central hub for cellular activity, driving cell identity and behavior. Despite its crucial role, understanding how biomaterials influence the nucleus remains an underexplored area of research. In our opinion, this is an overlooked opportunity, particularly in regenerative medicine - a field where cellular control is not just beneficial, but essential. As such, we emphasize the need to recognize nuclear characteristics as a key metric for evaluating material functionality. In this leading opinion article, we discuss how state-of-the-art technologies can help reveal biomaterial-driven nuclear alterations, offering crucial insights that will advance the field of regenerative medicine.
Collapse
Affiliation(s)
- Steven Vermeulen
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Elizabeth Rosado Balmayor
- Experimental Orthopaedics and Trauma Surgery, Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen, Germany
- Musculoskeletal Gene Therapy Laboratory, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
26
|
Salama A, Tolba E, Saleh AK, Cruz-Maya I, Alvarez-Perez MA, Guarino V. Biomineralization of Polyelectrolyte-Functionalized Electrospun Fibers: Optimization and In Vitro Validation for Bone Applications. Biomimetics (Basel) 2024; 9:253. [PMID: 38667264 PMCID: PMC11048701 DOI: 10.3390/biomimetics9040253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
In recent years, polyelectrolytes have been successfully used as an alternative to non-collagenous proteins to promote interfibrillar biomineralization, to reproduce the spatial intercalation of mineral phases among collagen fibrils, and to design bioinspired scaffolds for hard tissue regeneration. Herein, hybrid nanofibers were fabricated via electrospinning, by using a mixture of Poly ɛ-caprolactone (PCL) and cationic cellulose derivatives, i.e., cellulose-bearing imidazolium tosylate (CIMD). The obtained fibers were self-assembled with Sodium Alginate (SA) by polyelectrolyte interactions with CIMD onto the fiber surface and, then, treated with simulated body fluid (SBF) to promote the precipitation of calcium phosphate (CaP) deposits. FTIR analysis confirmed the presence of SA and CaP, while SEM equipped with EDX analysis mapped the calcium phosphate constituent elements, estimating an average Ca/P ratio of about 1.33-falling in the range of biological apatites. Moreover, in vitro studies have confirmed the good response of mesenchymal cells (hMSCs) on biomineralized samples, since day 3, with a significant improvement in the presence of SA, due to the interaction of SA with CaP deposits. More interestingly, after a decay of metabolic activity on day 7, a relevant increase in cell proliferation can be recognized, in agreement with the beginning of the differentiation phase, confirmed by ALP results. Antibacterial tests performed by using different bacteria populations confirmed that nanofibers with an SA-CIMD complex show an optimal inhibitory response against S. mutans, S. aureus, and E. coli, with no significant decay due to the effect of CaP, in comparison with non-biomineralized controls. All these data suggest a promising use of these biomineralized fibers as bioinspired membranes with efficient antimicrobial and osteoconductive cues suitable to support bone healing/regeneration.
Collapse
Affiliation(s)
- Ahmed Salama
- Cellulose and Paper Department, National Research Centre, 33 El Bohouth St., Dokki, Giza 12622, Egypt;
| | - Emad Tolba
- Polymers and Pigments Department, National Research Centre, 33 El-Buhouth St., Dokki, Giza 12622, Egypt
| | - Ahmed K. Saleh
- Cellulose and Paper Department, National Research Centre, 33 El Bohouth St., Dokki, Giza 12622, Egypt;
| | - Iriczalli Cruz-Maya
- Institute of Polymers, Composite and Biomaterials, National Research Council of Italy, Mostra d’Oltremare, V.le J.F. Kennedy 54, 80125 Naples, Italy;
| | - Marco A. Alvarez-Perez
- Tissue Bioengineering Laboratory, DEPeI, School of Dentistry, Universidad Nacional Autonoma de Mexico (UNAM), Circuito Exterior s/n C.P., Mexico City 04510, Mexico;
| | - Vincenzo Guarino
- Institute of Polymers, Composite and Biomaterials, National Research Council of Italy, Mostra d’Oltremare, V.le J.F. Kennedy 54, 80125 Naples, Italy;
| |
Collapse
|
27
|
Jia K, You J, Zhu Y, Li M, Chen S, Ren S, Chen S, Zhang J, Wang H, Zhou Y. Platelet-rich fibrin as an autologous biomaterial for bone regeneration: mechanisms, applications, optimization. Front Bioeng Biotechnol 2024; 12:1286035. [PMID: 38689760 PMCID: PMC11058865 DOI: 10.3389/fbioe.2024.1286035] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/22/2024] [Indexed: 05/02/2024] Open
Abstract
Platelet-rich fibrin, a classical autologous-derived bioactive material, consists of a fibrin scaffold and its internal loading of growth factors, platelets, and leukocytes, with the gradual degradation of the fibrin scaffold and the slow release of physiological doses of growth factors. PRF promotes vascular regeneration, promotes the proliferation and migration of osteoblast-related cells such as mesenchymal cells, osteoblasts, and osteoclasts while having certain immunomodulatory and anti-bacterial effects. PRF has excellent osteogenic potential and has been widely used in the field of bone tissue engineering and dentistry. However, there are still some limitations of PRF, and the improvement of its biological properties is one of the most important issues to be solved. Therefore, it is often combined with bone tissue engineering scaffolds to enhance its mechanical properties and delay its degradation. In this paper, we present a systematic review of the development of platelet-rich derivatives, the structure and biological properties of PRF, osteogenic mechanisms, applications, and optimization to broaden their clinical applications and provide guidance for their clinical translation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| |
Collapse
|
28
|
Tanvir MAH, Khaleque MA, Kim GH, Yoo WY, Kim YY. The Role of Bioceramics for Bone Regeneration: History, Mechanisms, and Future Perspectives. Biomimetics (Basel) 2024; 9:230. [PMID: 38667241 PMCID: PMC11048714 DOI: 10.3390/biomimetics9040230] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Osteoporosis is a skeletal disorder marked by compromised bone integrity, predisposing individuals, particularly older adults and postmenopausal women, to fractures. The advent of bioceramics for bone regeneration has opened up auspicious pathways for addressing osteoporosis. Research indicates that bioceramics can help bones grow back by activating bone morphogenetic protein (BMP), mitogen-activated protein kinase (MAPK), and wingless/integrated (Wnt)/β-catenin pathways in the body when combined with stem cells, drugs, and other supports. Still, bioceramics have some problems, such as not being flexible enough and prone to breaking, as well as difficulties in growing stem cells and discovering suitable supports for different bone types. While there have been improvements in making bioceramics better for healing bones, it is important to keep looking for new ideas from different areas of medicine to make them even better. By conducting a thorough scrutiny of the pivotal role bioceramics play in facilitating bone regeneration, this review aspires to propel forward the rapidly burgeoning domain of scientific exploration. In the end, this appreciation will contribute to the development of novel bioceramics that enhance bone regrowth and offer patients with bone disorders alternative treatments.
Collapse
Affiliation(s)
| | | | | | | | - Young-Yul Kim
- Department of Orthopedic Surgery, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Daejeon 34943, Republic of Korea; (M.A.H.T.); (M.A.K.); (G.-H.K.); (W.-Y.Y.)
| |
Collapse
|
29
|
Li M, Jin M, Yang H. Remodelers of the vascular microenvironment: The effect of biopolymeric hydrogels on vascular diseases. Int J Biol Macromol 2024; 264:130764. [PMID: 38462100 DOI: 10.1016/j.ijbiomac.2024.130764] [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: 12/19/2023] [Revised: 01/31/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
Vascular disease is the leading health problem worldwide. Vascular microenvironment encompasses diverse cell types, including those within the vascular wall, blood cells, stromal cells, and immune cells. Initiation of the inflammatory state of the vascular microenvironment and changes in its mechanics can profoundly affect vascular homeostasis. Biomedical materials play a crucial role in modern medicine, hydrogels, characterized by their high-water content, have been increasingly utilized as a three-dimensional interaction network. In recent times, the remarkable progress in utilizing hydrogels and understanding vascular microenvironment have enabled the treatment of vascular diseases. In this review, we give an emphasis on the utilization of hydrogels and their advantages in the various vascular diseases including atherosclerosis, aneurysm, vascular ulcers of the lower limbs and myocardial infarction. Further, we highlight the importance and advantages of hydrogels as artificial microenvironments.
Collapse
Affiliation(s)
- Minhao Li
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang 110122, Liaoning Province, China
| | - Meiqi Jin
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang 110122, Liaoning Province, China
| | - Huazhe Yang
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang 110122, Liaoning Province, China.
| |
Collapse
|
30
|
Zhao Y, Sun W, Wu X, Gao X, Song F, Duan B, Lu A, Yang H, Huang C. Janus Membrane with Intrafibrillarly Strontium-Apatite-Mineralized Collagen for Guided Bone Regeneration. ACS NANO 2024; 18:7204-7222. [PMID: 38373291 DOI: 10.1021/acsnano.3c12403] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Commercial collagen membranes face difficulty in guided bone regeneration (GBR) due to the absence of hierarchical structural design, effective interface management, and diverse bioactivity. Herein, a Janus membrane called SrJM is developed that consists of a porous collagen face to enhance osteogenic function and a dense face to maintain barrier function. Specifically, biomimetic intrafibrillar mineralization of collagen with strontium apatite is realized by liquid precursors of amorphous strontium phosphate. Polycaprolactone methacryloyl is further integrated on one side of the collagen as a dense face, which endows SrJM with mechanical support and a prolonged lifespan. In vitro experiments demonstrate that the dense face of SrJM acts as a strong barrier against fibroblasts, while the porous face significantly promotes cell adhesion and osteogenic differentiation through activation of calcium-sensitive receptor/integrin/Wnt signaling pathways. Meanwhile, SrJM effectively enhances osteogenesis and angiogenesis by recruiting stem cells and modulating osteoimmune response, thus creating an ideal microenvironment for bone regeneration. In vivo studies verify that the bone defect region guided by SrJM is completely repaired by newly formed vascularized bone. Overall, the outstanding performance of SrJM supports its ongoing development as a multifunctional GBR membrane, and this study provides a versatile strategy of fabricating collagen-based biomaterials for hard tissue regeneration.
Collapse
Affiliation(s)
- Yaning Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Wei Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Xiaoyi Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Xin Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Bo Duan
- Interdisciplinary Institute of NMR and Molecular Sciences, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Ang Lu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| |
Collapse
|
31
|
Yun C, Kim SH, Kim KM, Yang MH, Byun MR, Kim JH, Kwon D, Pham HTM, Kim HS, Kim JH, Jung YS. Advantages of Using 3D Spheroid Culture Systems in Toxicological and Pharmacological Assessment for Osteogenesis Research. Int J Mol Sci 2024; 25:2512. [PMID: 38473760 DOI: 10.3390/ijms25052512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Bone differentiation is crucial for skeletal development and maintenance. Its dysfunction can cause various pathological conditions such as rickets, osteoporosis, osteogenesis imperfecta, or Paget's disease. Although traditional two-dimensional cell culture systems have contributed significantly to our understanding of bone biology, they fail to replicate the intricate biotic environment of bone tissue. Three-dimensional (3D) spheroid cell cultures have gained widespread popularity for addressing bone defects. This review highlights the advantages of employing 3D culture systems to investigate bone differentiation. It highlights their capacity to mimic the complex in vivo environment and crucial cellular interactions pivotal to bone homeostasis. The exploration of 3D culture models in bone research offers enhanced physiological relevance, improved predictive capabilities, and reduced reliance on animal models, which have contributed to the advancement of safer and more effective strategies for drug development. Studies have highlighted the transformative potential of 3D culture systems for expanding our understanding of bone biology and developing targeted therapeutic interventions for bone-related disorders. This review explores how 3D culture systems have demonstrated promise in unraveling the intricate mechanisms governing bone homeostasis and responses to pharmacological agents.
Collapse
Affiliation(s)
- Chawon Yun
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Sou Hyun Kim
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Kyung Mok Kim
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Min Hye Yang
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Mi Ran Byun
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Joung-Hee Kim
- Department of Medical Beauty Care, Dongguk University Wise, Gyeongju 38066, Republic of Korea
| | - Doyoung Kwon
- Jeju Research Institute of Pharmaceutical Sciences, College of Pharmacy, Jeju National University, Jeju 63243, Republic of Korea
| | - Huyen T M Pham
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hyo-Sop Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Jae-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Young-Suk Jung
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
32
|
Sass JO, Sellin ML, Kauertz E, Johannsen J, Weinmann M, Stenzel M, Frank M, Vogel D, Bader R, Jonitz-Heincke A. Advanced Ti-Nb-Ta Alloys for Bone Implants with Improved Functionality. J Funct Biomater 2024; 15:46. [PMID: 38391899 PMCID: PMC10889793 DOI: 10.3390/jfb15020046] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024] Open
Abstract
The additive manufacturing of titanium-niobium-tantalum alloys with nominal chemical compositions Ti-xNb-6Ta (x = 20, 27, 35) by means of laser beam powder bed fusion is reported, and their potential as implant materials is elaborated by mechanical and biological characterization. The properties of dense specimens manufactured in different build orientations and of open porous Ti-20Nb-6Ta specimens are evaluated. Compression tests indicate that strength and elasticity are influenced by the chemical composition and build orientation. The minimum elasticity is always observed in the 90° orientation. It is lowest for Ti-20Nb-6Ta (43.2 ± 2.7 GPa) and can be further reduced to 8.1 ± 1.0 GPa for open porous specimens (p < 0.001). Furthermore, human osteoblasts are cultivated for 7 and 14 days on as-printed specimens and their biological response is compared to that of Ti-6Al-4V. Build orientation and cultivation time significantly affect the gene expression profile of osteogenic differentiation markers. Incomplete cell spreading is observed in specimens manufactured in 0° build orientation, whereas widely stretched cells are observed in 90° build orientation, i.e., parallel to the build direction. Compared to Ti-6Al-4V, Ti-Nb-Ta specimens promote improved osteogenesis and reduce the induction of inflammation. Accordingly, Ti-xNb-6Ta alloys have favorable mechanical and biological properties with great potential for application in orthopedic implants.
Collapse
Affiliation(s)
- Jan-Oliver Sass
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| | - Marie-Luise Sellin
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| | - Elisa Kauertz
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| | - Jan Johannsen
- Fraunhofer Research Institution for Additive Manufacturing Technologies IAPT, Am Schleusengraben 14, 21029 Hamburg, Germany
| | | | | | - Marcus Frank
- Medical Biology and Electron Microscopy Center, Rostock University Medical Center, Strempelstraße 14, 18057 Rostock, Germany
- Department Life, Light and Matter, University of Rostock, 18051 Rostock, Germany
| | - Danny Vogel
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| | - Rainer Bader
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
- Department Life, Light and Matter, University of Rostock, 18051 Rostock, Germany
| | - Anika Jonitz-Heincke
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| |
Collapse
|
33
|
Zhang Y, Li G, Wang J, Zhou F, Ren X, Su J. Small Joint Organoids 3D Bioprinting: Construction Strategy and Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302506. [PMID: 37814373 DOI: 10.1002/smll.202302506] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 09/28/2023] [Indexed: 10/11/2023]
Abstract
Osteoarthritis (OA) is a chronic disease that causes pain and disability in adults, affecting ≈300 million people worldwide. It is caused by damage to cartilage, including cellular inflammation and destruction of the extracellular matrix (ECM), leading to limited self-repairing ability due to the lack of blood vessels and nerves in the cartilage tissue. Organoid technology has emerged as a promising approach for cartilage repair, but constructing joint organoids with their complex structures and special mechanisms is still challenging. To overcome these boundaries, 3D bioprinting technology allows for the precise design of physiologically relevant joint organoids, including shape, structure, mechanical properties, cellular arrangement, and biological cues to mimic natural joint tissue. In this review, the authors will introduce the biological structure of joint tissues, summarize key procedures in 3D bioprinting for cartilage repair, and propose strategies for constructing joint organoids using 3D bioprinting. The authors also discuss the challenges of using joint organoids' approaches and perspectives on their future applications, opening opportunities to model joint tissues and response to joint disease treatment.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Trauma Orthopedics, Zhongye Hospital, Shanghai, 200941, China
| | - Jian Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Fengjin Zhou
- Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Xiaoxiang Ren
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
34
|
Han Z, Xiong J, Jin X, Dai Q, Han M, Wu H, Yang J, Tang H, He L. Advances in reparative materials for infectious bone defects and their applications in maxillofacial regions. J Mater Chem B 2024; 12:842-871. [PMID: 38173410 DOI: 10.1039/d3tb02069j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Infectious bone defects are characterized by the partial loss or destruction of bone tissue resulting from bacterial contaminations subsequent to diseases or external injuries. Traditional bone transplantation and clinical methods are insufficient in meeting the treatment demands for such diseases. As a result, researchers have increasingly focused on the development of more sophisticated biomaterials for improved therapeutic outcomes in recent years. This review endeavors to investigate specific reparative materials utilized for the treatment of infectious bone defects, particularly those present in the maxillofacial region, with a focus on biomaterials capable of releasing therapeutic substances, functional contact biomaterials, and novel physical therapy materials. These biomaterials operate via heightened antibacterial or osteogenic properties in order to eliminate bacteria and/or stimulate bone cells regeneration in the defect, ultimately fostering the reconstitution of maxillofacial bone tissue. Based upon some successful applications of new concept materials in bone repair of other parts, we also explore their future prospects and potential uses in maxillofacial bone repair later in this review. We highlight that the exploration of advanced biomaterials holds promise in establishing a solid foundation for the development of more biocompatible, effective, and personalized treatments for reconstructing infectious maxillofacial defects.
Collapse
Affiliation(s)
- Ziyi Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jingdi Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Xiaohan Jin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Qinyue Dai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Mingyue Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Hongkun Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jiaojiao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Haiqin Tang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Libang He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
35
|
Xu L, Jacobs R, Cao Y, Sun X, Qin X. Tissue-engineered bone construct promotes early osseointegration of implants with low primary stability in oversized osteotomy. BMC Oral Health 2024; 24:69. [PMID: 38200461 PMCID: PMC10782778 DOI: 10.1186/s12903-023-03834-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
OBJECTIVES To evaluate the histological parameters and bone mechanical properties around implants with low primary stability (PS) in grafted bone substitutes within an oversized osteotomy. MATERIALS AND METHODS An oversized osteotomy penetrating the double cortical bone layers was made on both femora of 24 New Zealand white rabbits. Bilaterally in the femur of all animals, 48 implants were installed, subdivided into four groups, corresponding to four prepared tissue-engineering bone complexes (TEBCs), which were placed between the implant surface and native bone wall: A: tricalcium phosphate β (TCP-β); B: autologous adipose derived-stem cells with TCP-β (ASCs/TCP-β); C: ASCs transfected with the enhanced-GFP gene with TCP-β (EGFP-ASCs/TCP-β); D: ASCs transfected with the BMP-2 gene with TCP-β (BMP2-ASCs/TCP-β). Trichrome fluorescent labeling was conducted. Animals were sacrificed after eight weeks. The trichromatic fluorescent labeling (%TFL), area of new bone (%NB), residual material (%RM), bone-implant contact (%BIC), and the removal torque force (RTF, N/cm) were assessed. RESULTS ASCs were successfully isolated from adipose tissue, and the primary ASCs were induced into osteogenic, chondrogenic, and adipogenic differentiation. The BMP-2 overexpression of ASCs sustained for ten days and greatly enhanced the expression of osteopontin (OPN). At eight weeks post-implantation, increased %NB and RTF were found in all groups. The most significant value of %TFL, %BIC and lowest %RM was detected in group D. CONCLUSION The low PS implants osseointegrate with considerable new bone in grafted TEBCs within an oversized osteotomy. Applying BMP-2 overexpressing ASCs-based TEBC promoted earlier osseointegration and more solid bone mechanical properties on low PS implants. Bone graft offers a wedging effect for the implant with low PS at placement and promotes osteogenesis on their surface in the healing period.
Collapse
Affiliation(s)
- Lianyi Xu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, Hubei, China
- Department of Imaging and Pathology, OMFS-IMPATH, KU Leuven, Kapucijnenvoer 7, Leuven, 3000, Belgium
| | - Reinhilde Jacobs
- Department of Imaging and Pathology, OMFS-IMPATH, KU Leuven, Kapucijnenvoer 7, Leuven, 3000, Belgium
- Department of Dental Medicine, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Yingguang Cao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, Hubei, China
| | - Xiaojuan Sun
- Department of Oral and Maxillofacial Surgery, General Hospital, Ningxia Medical University, 804 Shengli Street, Yinchuan, 750004, China.
| | - Xu Qin
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, Hubei, China.
| |
Collapse
|
36
|
ISHIHARA K. Biomimetic polymers with phosphorylcholine groups as biomaterials for medical devices. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2024; 100:579-606. [PMID: 39662944 PMCID: PMC11704457 DOI: 10.2183/pjab.100.037] [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: 06/15/2024] [Accepted: 10/01/2024] [Indexed: 12/13/2024]
Abstract
Biomimetic molecular designs can yield superior biomaterials. Polymers with a phosphorylcholine group, a polar group of phospholipid molecules, are particularly interesting. A methacrylate monomer, 2-methacryloyloxyethyl phosphorylcholine (MPC), was developed using efficient synthetic reactions and purification techniques. This process has been applied in industrial production to supply MPC globally. Polymers with various structures can be readily synthesized using MPC and their properties have been studied. The MPC polymer surface has a highly hydrated structure in biological conditions, leading to the prevention of adsorption of proteins and lipid molecules, adhesion of cells, and inhibition of bacterial adhesion and biofilm formation. Additionally, it provides an extremely lubricious surface. MPC polymers are used in various applications and can be stably immobilized on material surfaces such as metals and ceramics and polymers such as elastomers. They are also stable under sterilization and in vivo conditions. This makes them ideal for application in the surface treatment of various medical devices, including artificial organs, implanted in humans.
Collapse
Affiliation(s)
- Kazuhiko ISHIHARA
- Division of Materials & Manufacturing Science, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
37
|
Eichholz KF, Pitacco P, Burdis R, Chariyev‐Prinz F, Barceló X, Tornifoglio B, Paetzold R, Garcia O, Kelly DJ. Integrating Melt Electrowriting and Fused Deposition Modeling to Fabricate Hybrid Scaffolds Supportive of Accelerated Bone Regeneration. Adv Healthc Mater 2024; 13:e2302057. [PMID: 37933556 PMCID: PMC11468945 DOI: 10.1002/adhm.202302057] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/23/2023] [Indexed: 11/08/2023]
Abstract
Emerging additive manufacturing (AM) strategies can enable the engineering of hierarchal scaffold structures for guiding tissue regeneration. Here, the advantages of two AM approaches, melt electrowriting (MEW) and fused deposition modelling (FDM), are leveraged and integrated to fabricate hybrid scaffolds for large bone defect healing. MEW is used to fabricate a microfibrous core to guide bone healing, while FDM is used to fabricate a stiff outer shell for mechanical support, with constructs being coated with pro-osteogenic calcium phosphate (CaP) nano-needles. Compared to MEW scaffolds alone, hybrid scaffolds prevent soft tissue collapse into the defect region and support increased vascularization and higher levels of new bone formation 12 weeks post-implantation. In an additional group, hybrid scaffolds are also functionalized with BMP2 via binding to the CaP coating, which further accelerates healing and facilitates the complete bridging of defects after 12 weeks. Histological analyses demonstrate that such scaffolds support the formation of well-defined annular bone, with an open medullary cavity, smooth periosteal surface, and no evidence of abnormal ectopic bone formation. These results demonstrate the potential of integrating different AM approaches for the development of regenerative biomaterials, and in particular, demonstrate the enhanced bone healing outcomes possible with hybrid MEW-FDM constructs.
Collapse
Affiliation(s)
- Kian F. Eichholz
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College Dublin152‐160 Pearse StreetDublinD02 R590Ireland
- Department of Mechanical, Manufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 VH29Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 CP49Ireland
| | - Pierluca Pitacco
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College Dublin152‐160 Pearse StreetDublinD02 R590Ireland
- Department of Mechanical, Manufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 VH29Ireland
| | - Ross Burdis
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College Dublin152‐160 Pearse StreetDublinD02 R590Ireland
- Department of Mechanical, Manufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 VH29Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 CP49Ireland
| | - Farhad Chariyev‐Prinz
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College Dublin152‐160 Pearse StreetDublinD02 R590Ireland
- Department of Mechanical, Manufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 VH29Ireland
| | - Xavier Barceló
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College Dublin152‐160 Pearse StreetDublinD02 R590Ireland
- Department of Mechanical, Manufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 VH29Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 CP49Ireland
| | - Brooke Tornifoglio
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College Dublin152‐160 Pearse StreetDublinD02 R590Ireland
- Department of Mechanical, Manufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 VH29Ireland
| | - Ryan Paetzold
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 CP49Ireland
- School of Mechanical and Materials EngineeringUniversity College DublinDublinD04 E4X0Ireland
| | - Orquidea Garcia
- Johnson & Johnson 3D Printing Innovation and Customer SolutionsJohnson & Johnson Services, Inc.IrvineCA92618USA
| | - Daniel J Kelly
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College Dublin152‐160 Pearse StreetDublinD02 R590Ireland
- Department of Mechanical, Manufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 VH29Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 CP49Ireland
- Department of Anatomy and Regenerative MERoyal College of Surgeons in IrelandDublinD02 YN77Ireland
| |
Collapse
|
38
|
Yuan S, Yang X, Wang X, Chen J, Tian W, Yang B. Injectable Xenogeneic Dental Pulp Decellularized Extracellular Matrix Hydrogel Promotes Functional Dental Pulp Regeneration. Int J Mol Sci 2023; 24:17483. [PMID: 38139310 PMCID: PMC10743504 DOI: 10.3390/ijms242417483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
The present challenge in dental pulp tissue engineering scaffold materials lies in the development of tissue-specific scaffolds that are conducive to an optimal regenerative microenvironment and capable of accommodating intricate root canal systems. This study utilized porcine dental pulp to derive the decellularized extracellular matrix (dECM) via appropriate decellularization protocols. The resultant dECM was dissolved in an acid pepsin solution to form dECM hydrogels. The analysis encompassed evaluating the microstructure and rheological properties of dECM hydrogels and evaluated their biological properties, including in vitro cell viability, proliferation, migration, tube formation, odontogenic, and neurogenic differentiation. Gelatin methacrylate (GelMA) hydrogel served as the control. Subsequently, hydrogels were injected into treated dentin matrix tubes and transplanted subcutaneously into nude mice to regenerate dental pulp tissue in vivo. The results showed that dECM hydrogels exhibited exceptional injectability and responsiveness to physiological temperature. It supported the survival, odontogenic, and neurogenic differentiation of dental pulp stem cells in a 3D culture setting. Moreover, it exhibited a superior ability to promote cell migration and angiogenesis compared to GelMA hydrogel in vitro. Additionally, the dECM hydrogel demonstrated the capability to regenerate pulp-like tissue with abundant blood vessels and a fully formed odontoblast-like cell layer in vivo. These findings highlight the potential of porcine dental pulp dECM hydrogel as a specialized scaffold material for dental pulp regeneration.
Collapse
Affiliation(s)
- Shengmeng Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (S.Y.); (X.W.); (J.C.)
- National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xueting Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (S.Y.); (X.W.); (J.C.)
- National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiuting Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (S.Y.); (X.W.); (J.C.)
- National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jinlong Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (S.Y.); (X.W.); (J.C.)
- National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (S.Y.); (X.W.); (J.C.)
- National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bo Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (S.Y.); (X.W.); (J.C.)
- National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
39
|
Yu Z, Wang H, Ying B, Mei X, Zeng D, Liu S, Qu W, Pan X, Pu S, Li R, Qin Y. Mild photothermal therapy assist in promoting bone repair: Related mechanism and materials. Mater Today Bio 2023; 23:100834. [PMID: 38024841 PMCID: PMC10643361 DOI: 10.1016/j.mtbio.2023.100834] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/21/2023] [Accepted: 10/14/2023] [Indexed: 12/01/2023] Open
Abstract
Achieving precision treatment in bone tissue engineering (BTE) remains a challenge. Photothermal therapy (PTT), as a form of precision therapy, has been extensively investigated for its safety and efficacy. It has demonstrated significant potential in the treatment of orthopedic diseases such as bone tumors, postoperative infections and osteoarthritis. However, the high temperatures associated with PTT can lead to certain limitations and drawbacks. In recent years, researchers have explored the use of biomaterials for mild photothermal therapy (MPT), which offers a promising approach for addressing these limitations. This review provides a comprehensive overview of the mechanisms underlying MPT and presents a compilation of photothermal agents and their utilization strategies for bone tissue repair. Additionally, the paper discusses the future prospects of MPT-assisted bone tissue regeneration, aiming to provide insights and recommendations for optimizing material design in this field.
Collapse
Affiliation(s)
- Zehao Yu
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Hao Wang
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Boda Ying
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Xiaohan Mei
- National & Local Joint Engineering Laboratory for Synthesis Technology of High-Performance Polymer, College of Chemistry, Jilin University, Changchun, 130012, People’s Republic of China
| | - Dapeng Zeng
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Shibo Liu
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Wenrui Qu
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Xiangjun Pan
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Si Pu
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Ruiyan Li
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| | - Yanguo Qin
- Department of Joint Surgery of Orthopaedic Center, The Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Jilin Provincial Key Laboratory of Orhtopeadics, Changchun, Jilin 130041 People’s Republic of China
| |
Collapse
|
40
|
Liu C, Zhang X, Zhao L, Hui L, Liu D. Multilayer amnion-PCL nanofibrous membrane loaded with celecoxib exerts a therapeutic effect against tendon adhesion by improving the inflammatory microenvironment. Heliyon 2023; 9:e23214. [PMID: 38144330 PMCID: PMC10746461 DOI: 10.1016/j.heliyon.2023.e23214] [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: 04/09/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 12/26/2023] Open
Abstract
Tendon adhesion is a common complication after tendon surgery. The inflammatory phase of tendon healing is characterized by the release of a large number of inflammatory factors, whose mediated excessive inflammatory response is an important cause of tendon adhesion formation. Nonsteroidal anti-inflammatory drugs(NSAIDs) were used to prevent tendon adhesions by reducing the inflammatory response. However, recent studies have shown that the NSAIDs partially impairs tendon healing. Therefore, optimizing the anti-adhesive membrane loaded with NSAIDs to mitigate the effects on tendon healing requires further in-depth study. Amniotic membranes(AM) are natural polymeric semi-permeable membranes from living organisms that are rich in matrix, growth factors, and other active ingredients. In this study, we used electrostatic spinning technology to construct multifunctional nanofiber membranes of the PCL membrane loaded with celecoxib and AM. In vitro cellular assays revealed that celecoxib-loaded PCL membranes significantly inhibited the adhesion and proliferation of fibroblasts with increasing concentrations of celecoxib. In a rabbit tendon repair model, biomechanical tests further confirmed that the PCL membrane loaded with celecoxib had better anti-adhesion effects. Further experimental studies revealed that the PCL/AM membrane improved the inflammatory microenvironment by downregulating the expression of pro-inflammatory factors such as COX-2, IL-1β, and TNF-α proteins; and inhibiting the synthesis of COL I and COL Ⅲ. The PCL/AM membrane can continuously release celecoxib to reduce the inflammatory response and deliver growth factors to the damaged area to build a suitable microenvironment for tendon repair, which provides a new direction to improve the repair efficiency of tendon.
Collapse
Affiliation(s)
- Chunjie Liu
- Xingtai People's Hospital Postdoctoral Workstation, Xingtai People's Hospital, No.16, Hongxing Street, Xingtai 054031, China
- Postdoctoral Mobile Station, Hebei Medical University, No.361, Zhongshan Road, Shijiazhuang 050017, China
- Department of Orthopedics, Tangshan Workers Hospital, No.27, Wenhua Road, Tangshan 063000, China
| | - Xiaochong Zhang
- Department of Research and Education, Xingtai People's Hospital, No.16, Hongxing Street, Xingtai 054031, China
| | - Lili Zhao
- Xingtai People's Hospital Postdoctoral Workstation, Xingtai People's Hospital, No.16, Hongxing Street, Xingtai 054031, China
- Department of Orthopedics, Xingtai People's Hospital, No.16, Hongxing Street, Xingtai 054031, China
| | - Limin Hui
- Department of Gynecology, Xingtai People's Hospital, No.16, Hongxing Street, Xingtai 054001, China
| | - Dengxiang Liu
- Institute of Cancer Control, Xingtai People's Hospital, No.16, Hongxing Street, Xingtai 054001, China
- Xingtai Key Laboratory of Precision Medicine for Liver Cirrhosis and Portal Hypertension, Xingtai People's Hospital, No.16, Hongxing Street, Xingtai 054001, China
| |
Collapse
|
41
|
Ghasemzadeh-Hasankolaei M, Pinto CA, Jesus D, Saraiva JA, Mano JF. Effect of high cyclic hydrostatic pressure on osteogenesis of mesenchymal stem cells cultured in liquefied micro-compartments. Mater Today Bio 2023; 23:100861. [PMID: 38058695 PMCID: PMC10696388 DOI: 10.1016/j.mtbio.2023.100861] [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: 02/23/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023] Open
Abstract
Bone resident cells are constantly subjected to a range of distinct mechanical loadings, which generates a complex microenvironment. In particular, hydrostatic pressure (HP) has a key impact on modulation of cell function and fate determination. Although HP is a constant mechanical stimulus, its role in regulating the osteogenesis process within a defined 3D microenvironment has not been comprehensively elucidated. Perceiving how environmental factors regulate the differentiation of stem cells is essential for expanding their regenerative potential. Inspired by the mechanical environment of bone, this study attempted to investigate the influence of different ranges of cyclic HP on human adipose-derived mesenchymal stem cells (MSCs) encapsulated within a compartmentalized liquefied microenvironment. Taking advantage of the liquefied environment of microcapsules, MSCs were exposed to cyclic HP of 5 or 50 MPa, 3 times/week at 37 °C. Biological tests using fluorescence staining of F-actin filaments showed a noticeable improvement in cell-cell interactions and cellular network formation of MSCs. These observations were more pronounced in osteogenic (OST) condition, as confirmed by fluorescent staining of vinculin. More interestingly, there was a significant increase in alkaline phosphatase activity of MSCs exposed to 50 MPa magnitude of HP, even in the absence of osteoinductive factors. In addition, a greater staining area of both osteopontin and hydroxyapatite was detected in the 50 MPa/OST group. These findings highlight the benefit of hydrostatic pressure to regulate osteogenesis of MSCs as well as the importance of employing simultaneous biochemical and mechanical stimulation to accelerate the osteogenic potential of MSCs for biomedical purposes.
Collapse
Affiliation(s)
| | - Carlos A. Pinto
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Diana Jesus
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Jorge A. Saraiva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - João F. Mano
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| |
Collapse
|
42
|
Vermeulen S, Knoops K, Duimel H, Parvizifard M, van Beurden D, López-Iglesias C, Giselbrecht S, Truckenmüller R, Habibović P, Tahmasebi Birgani Z. An in vitro model system based on calcium- and phosphate ion-induced hMSC spheroid mineralization. Mater Today Bio 2023; 23:100844. [PMID: 38033367 PMCID: PMC10682137 DOI: 10.1016/j.mtbio.2023.100844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
A challenge in regenerative medicine is creating the three-dimensional organic and inorganic in vitro microenvironment of bone, which would allow the study of musculoskeletal disorders and the generation of building blocks for bone regeneration. This study presents a microwell-based platform for creating spheroids of human mesenchymal stromal cells, which are then mineralized using ionic calcium and phosphate supplementation. The resulting mineralized spheroids promote an osteogenic gene expression profile through the influence of the spheroids' biophysical environment and inorganic signaling and require less calcium or phosphate to achieve mineralization compared to a monolayer culture. We found that mineralized spheroids represent an in vitro model for studying small molecule perturbations and extracellular mediated calcification. Furthermore, we demonstrate that understanding pathway signaling elicited by the spheroid environment allows mimicking these pathways in traditional monolayer culture, enabling similar rapid mineralization events. In sum, this study demonstrates the rapid generation and employment of a mineralized cell model system for regenerative medicine applications.
Collapse
Affiliation(s)
- Steven Vermeulen
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Kèvin Knoops
- Microscopy CORE Lab, M4I Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Hans Duimel
- Microscopy CORE Lab, M4I Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Maryam Parvizifard
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Denis van Beurden
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Carmen López-Iglesias
- Microscopy CORE Lab, M4I Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Zeinab Tahmasebi Birgani
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
43
|
Luo Q, Shang K, Zhu J, Wu Z, Cao T, Ahmed AAQ, Huang C, Xiao L. Biomimetic cell culture for cell adhesive propagation for tissue engineering strategies. MATERIALS HORIZONS 2023; 10:4662-4685. [PMID: 37705440 DOI: 10.1039/d3mh00849e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Biomimetic cell culture, which involves creating a biomimetic microenvironment for cells in vitro by engineering approaches, has aroused increasing interest given that it maintains the normal cellular phenotype, genotype and functions displayed in vivo. Therefore, it can provide a more precise platform for disease modelling, drug development and regenerative medicine than the conventional plate cell culture. In this review, initially, we discuss the principle of biomimetic cell culture in terms of the spatial microenvironment, chemical microenvironment, and physical microenvironment. Then, the main strategies of biomimetic cell culture and their state-of-the-art progress are summarized. To create a biomimetic microenvironment for cells, a variety of strategies has been developed, ranging from conventional scaffold strategies, such as macroscopic scaffolds, microcarriers, and microgels, to emerging scaffold-free strategies, such as spheroids, organoids, and assembloids, to simulate the native cellular microenvironment. Recently, 3D bioprinting and microfluidic chip technology have been applied as integrative platforms to obtain more complex biomimetic structures. Finally, the challenges in this area are discussed and future directions are discussed to shed some light on the community.
Collapse
Affiliation(s)
- Qiuchen Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Keyuan Shang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Jing Zhu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Zhaoying Wu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Tiefeng Cao
- Department of Gynaecology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510070, China
| | - Abeer Ahmed Qaed Ahmed
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, 27100 Pavia, Italy
| | - Chixiang Huang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|
44
|
Lin H, Zhang L, Zhang Q, Wang Q, Wang X, Yan G. Mechanism and application of 3D-printed degradable bioceramic scaffolds for bone repair. Biomater Sci 2023; 11:7034-7050. [PMID: 37782081 DOI: 10.1039/d3bm01214j] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Bioceramics have attracted considerable attention in the field of bone repair because of their excellent osteogenic properties, degradability, and biocompatibility. To resolve issues regarding limited formability, recent studies have introduced 3D printing technology for the fabrication of bioceramic bone repair scaffolds. Nevertheless, the mechanisms by which bioceramics promote bone repair and clinical applications of 3D-printed bioceramic scaffolds remain elusive. This review provides an account of the fabrication methods of 3D-printed degradable bioceramic scaffolds. In addition, the types and characteristics of degradable bioceramics used in clinical and preclinical applications are summarized. We have also highlighted the osteogenic molecular mechanisms in biomaterials with the aim of providing a basis and support for future research on the clinical applications of degradable bioceramic scaffolds. Finally, new developments and potential applications of 3D-printed degradable bioceramic scaffolds are discussed with reference to experimental and theoretical studies.
Collapse
Affiliation(s)
- Hui Lin
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Liyun Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Qiyue Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Xue Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
| | - Guangqi Yan
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
| |
Collapse
|
45
|
Teimoori M, Nokhbatolfoghahaei H, Khojasteh A. Bilayer scaffolds/membranes for bone tissue engineering applications: A systematic review. BIOMATERIALS ADVANCES 2023; 153:213528. [PMID: 37352742 DOI: 10.1016/j.bioadv.2023.213528] [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: 04/09/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
OBJECTIVE This systematic review evaluates the purpose, materials, physio-mechanical, and biological effects of bilayer scaffolds/membranes used for bone tissue engineering applications. METHODS A comprehensive electronic search of English-language literature from 2012 to October 2022 was conducted in PubMed, Scopus, ScienceDirect, and Google Scholar online databases according to the PRISMA 2020 guidelines. The quality of animal studies was evaluated through the SYRCLE's risk of bias tool. RESULTS A total of 77 studies were sought for retrieval, and 39 studies met the inclusion criteria. According to the synthesis results, most bilayers had a dense barrier layer that prevented connective tissue penetration and a loose osteogenic layer that supported cell migration and osteogenesis. PLGA, PCL, and chitosan were the most common polymers in the barrier layers, while the most utilized polymers in osteogenic layers were PLGA and gelatin. Electrospinning and solvent casting were the most common fabrication methods to design the bilayer structures. Many studies reported higher biological results for bilayers compared to their single layers. Also, fabricated bilayers' in vitro osteogenesis and in vivo new bone formation were significantly superior or at least comparable to the frequently used commercial membranes. CONCLUSION 1) Bilayers with two distinct layers and different materials, porosities, mechanical properties, and biological behavior can significantly improve heterogeneous bone regeneration; 2) the addition of ceramics and/or drugs to the osteogenic layer enhances the osteogenic properties of the bilayers; 3) fabrication method and pore size of the layers play an important role in determining the mechanical and biological behavior of them.
Collapse
Affiliation(s)
- Mahdis Teimoori
- Student Research Committee, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Khojasteh
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Cranio-Maxillofacial Surgery, University Hospital, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
46
|
Zheng J, Wang Y, Wang Y, Duan R, Liu L. Gelatin/Hyaluronic Acid Photocrosslinked Double Network Hydrogel with Nano-Hydroxyapatite Composite for Potential Application in Bone Repair. Gels 2023; 9:742. [PMID: 37754423 PMCID: PMC10530748 DOI: 10.3390/gels9090742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/28/2023] Open
Abstract
The application of hydrogels in bone repair is limited due to their low mechanical strength. Simulating bone extracellular matrix, methylacrylylated gelatin (GelMA)/methylacrylylated hyaluronic acid (HAMA)/nano-hydroxyapatite(nHap) composite hydrogels were prepared by combining the double network strategy and composite of nHap in this study. The precursor solutions of the composite hydrogels were injectable due to their shear thinning property. The compressive elastic modulus of the composite hydrogel was significantly enhanced, the fracture strength of the composite hydrogel nearly reached 1 MPa, and the composite hydrogel retained its high water content at above 88%. The composite hydrogels possess good compatibility with BMSCS and have the potential to be used as injectable hydrogels for bone defect treatment.
Collapse
Affiliation(s)
| | | | | | | | - Lingrong Liu
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China; (J.Z.); (Y.W.); (Y.W.); (R.D.)
| |
Collapse
|
47
|
Iqbal S, Sohail M, Fang S, Ding J, Shen L, Chen M, Shu G, Du YZ, Ji J. Biomaterials evolution: from inert to instructive. Biomater Sci 2023; 11:6109-6115. [PMID: 37591802 DOI: 10.1039/d3bm00322a] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
The field of biomaterials has experienced substantial evolution in recent years, driven by advancements in materials science and engineering. This has led to an expansion of the biomaterials definition to include biocompatibility, bioactivity, bioderived materials, and biological tissues. Consequently, the intended performance of biomaterials has shifted from a passive role wherein a biomaterial is merely accepted by the body to an active role wherein a biomaterial instructs its biological environment. In the future, the integration of bioinspired designs and dynamic behavior into fabrication technologies will revolutionize the field of biomaterials. This perspective presents the recent advances in the evolution of biomaterials in fabrication technologies and provides a brief insight into smart biomaterials.
Collapse
Affiliation(s)
- Sajid Iqbal
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Muhammad Sohail
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Shiji Fang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Jiayi Ding
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Lin Shen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Gaofeng Shu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| |
Collapse
|
48
|
de Kanter AFJ, Jongsma KR, Bouten CVC, Bredenoord AL. How Smart are Smart Materials? A Conceptual and Ethical Analysis of Smart Lifelike Materials for the Design of Regenerative Valve Implants. SCIENCE AND ENGINEERING ETHICS 2023; 29:33. [PMID: 37668955 PMCID: PMC10480256 DOI: 10.1007/s11948-023-00453-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 08/09/2023] [Indexed: 09/06/2023]
Abstract
It may soon become possible not just to replace, but to re-grow healthy tissues after injury or disease, because of innovations in the field of Regenerative Medicine. One particularly promising innovation is a regenerative valve implant to treat people with heart valve disease. These implants are fabricated from so-called 'smart', 'lifelike' materials. Implanted inside a heart, these implants stimulate re-growth of a healthy, living heart valve. While the technological development advances, the ethical implications of this new technology are still unclear and a clear conceptual understanding of the notions 'smart' and 'lifelike' is currently lacking. In this paper, we explore the conceptual and ethical implications of the development of smart lifelike materials for the design of regenerative implants, by analysing heart valve implants as a showcase. In our conceptual analysis, we show that the materials are considered 'smart' because they can communicate with human tissues, and 'lifelike' because they are structurally similar to these tissues. This shows that regenerative valve implants become intimately integrated in the living tissues of the human body. As such, they manifest the ontological entanglement of body and technology. In our ethical analysis, we argue this is ethically significant in at least two ways: It exacerbates the irreversibility of the implantation procedure, and it might affect the embodied experience of the implant recipient. With our conceptual and ethical analysis, we aim to contribute to responsible development of smart lifelike materials and regenerative implants.
Collapse
Affiliation(s)
- Anne-Floor J de Kanter
- Department of Bioethics and Health Humanities, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3508 GA, Utrecht, The Netherlands.
| | - Karin R Jongsma
- Department of Bioethics and Health Humanities, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3508 GA, Utrecht, The Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| | - Annelien L Bredenoord
- Department of Bioethics and Health Humanities, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3508 GA, Utrecht, The Netherlands
- Erasmus School of Philosophy, Erasmus University Rotterdam, 3062 PA, Rotterdam, The Netherlands
| |
Collapse
|
49
|
Zhao C, Shu C, Yu J, Zhu Y. Metal-organic frameworks functionalized biomaterials for promoting bone repair. Mater Today Bio 2023; 21:100717. [PMID: 37545559 PMCID: PMC10401359 DOI: 10.1016/j.mtbio.2023.100717] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Bone defects induced by bone trauma, tumors and osteoarthritis greatly affect the life quality and health of patients. The biomaterials with numerous advantages are becoming the most preferred options for repairing bone defects and treating orthopedic diseases. However, their repairing effects remains unsatisfactory, especially in bone defects suffering from tumor, inflammation, and/or bacterial infection. There are several strategies to functionalize biomaterials, but a more general and efficient method is essential for accomplishing the functionalization of biomaterials. Possessing high specific surface, high porosity, controlled degradability and variable composition, metal-organic frameworks (MOFs) materials are inherently advantageous for functionalizing biomaterials, with tremendous improvements having been achieved. This review summarizes recent progresses in MOFs functionalized biomaterials for promoting bone repair and therapeutic effects. In specific, by utilizing various properties of diverse MOFs materials, integrated MOFs functionalized biomaterials achieve enhanced bone regeneration, antibacterial, anti-inflammatory and anti-tumor functions. Finally, the summary and prospects of on the development of MOFs-functionalized biomaterials for promoting bone repair were discussed.
Collapse
Affiliation(s)
- Chaoqian Zhao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Chaoqin Shu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Jiangming Yu
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiaotong University, Shanghai, 200336, PR China
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
50
|
He Y, Liang L, Luo C, Zhang ZY, Huang J. Strategies for in situ tissue engineering of vascularized bone regeneration (Review). Biomed Rep 2023; 18:42. [PMID: 37325184 PMCID: PMC10265129 DOI: 10.3892/br.2023.1625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/03/2023] [Indexed: 06/17/2023] Open
Abstract
Numerous physiological processes occur following bone fracture, including inflammatory cell recruitment, vascularization, and callus formation and remodeling. In particular circumstances, such as critical bone defects or osteonecrosis, the regenerative microenvironment is compromised, rendering endogenous stem/progenitor cells incapable of fully manifesting their reparative potential. Consequently, external interventions, such as grafting or augmentation, are frequently necessary. In situ bone tissue engineering (iBTE) employs cell-free scaffolds that possess microenvironmental cues, which, upon implantation, redirect the behavior of endogenous stem/progenitor cells towards a pro-regenerative inflammatory response and reestablish angiogenesis-osteogenesis coupling. This process ultimately results in vascularized bone regeneration (VBR). In this context, a comprehensive review of the current techniques and modalities in VBR-targeted iBTE technology is provided.
Collapse
Affiliation(s)
- Yijun He
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Lin Liang
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| | - Cheng Luo
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| | - Zhi-Yong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Jiongfeng Huang
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| |
Collapse
|