1
|
Liu Y, Zhou Z, Lu G, Zhang X, Shi D, Tong L, Chen D, Tuan RS, Li ZA. Musculoskeletal organoids: An emerging toolkit for establishing personalized models of musculoskeletal disorders and developing regenerative therapies. Acta Biomater 2025:S1742-7061(25)00362-9. [PMID: 40381929 DOI: 10.1016/j.actbio.2025.05.037] [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: 01/01/2025] [Revised: 05/09/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Musculoskeletal (MSK) conditions are the primary cause of physical disability globally. These disorders are physically and mentally debilitating and severely impact the patients' quality of life. As the median age of the world's population increases, there has been an intensifying urgency of developing efficacious therapies for various orthopaedic conditions. Furthermore, the highly heterogeneous nature of MSK conditions calls for a personalized approach to studying disease mechanisms and developing regenerative treatments. Organoids have emerged as an advanced approach to generating functional tissue/organ mimics in vitro, which hold promise in MSK regeneration, disease modeling, and therapeutic development. Herein, we review the preparation, characterization, and application of various MSK organoids. We highlight the potential of patient-specific organoids in the development of personalized medicine and discuss the challenges and opportunities in the future development of MSK organoids. STATEMENT OF SIGNIFICANCE: Despite decades of research, translation of MSK research into clinical applications remains limited, partially attributed to our inadequate understanding of disease mechanisms. To advance therapeutic development, there are critical needs for MSK disease models with higher clinical relevance and predictive power. Additionally, engineered constructs that closely mimic the structural and functional features of native MSK tissues are highly desirable. MSK organoids have emerged as a promising approach to meet the above requirements. To unleash the full potential of MSK organoids necessitates a comprehensive understanding of their categories, construction, development, functions, applications, and challenges. This review aims to fulfill this crucial need, aiming to accelerate the clinical translation of MSK organoid platforms to benefit millions of patients afflicted with MSK conditions.
Collapse
Affiliation(s)
- Yuwei Liu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China; Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, PR China
| | - Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Gang Lu
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong Special Administrative Region of China
| | - Xin Zhang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191 PR China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China
| | - Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; Department of Pharmacology, Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen 518000, PR China.
| | - Rocky S Tuan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong Special Administrative Region of China.
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong Special Administrative Region of China; Peter Hung Pain Research Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China.
| |
Collapse
|
2
|
Shi Y, Zhuo R, Guo W, Shi Y. A Quantitative Evaluation of the Efficacy of Endochondral Ossification-Based Grafts in Bone Defect Regeneration: An Analysis of Animal Studies. ACS Biomater Sci Eng 2025; 11:2481-2491. [PMID: 40301001 DOI: 10.1021/acsbiomaterials.4c01895] [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: 05/01/2025]
Abstract
The regeneration of bone defects through bone grafts primarily depends on two strategies: intramembrane ossification (IO) and endochondral ossification (EO). Traditional bone tissue engineering has focused on mimicking the IO process to stimulate the formation of a bone-like matrix. However, repair strategies based on IO often result in excessive deposition of the matrix on the graft surface, hindering bone tissue regeneration. In recent years, researchers have increasingly focused on investigating the reparative potential of EO-based grafts for bone defects, such as microspheres, pellets, and hydrogel. However, the effectiveness of EO-based grafts on bone defects has not yet been quantitatively evaluated. Therefore, this study conducted a systematic review and meta-analysis of previous studies to quantitatively assess the bone regenerative potential of EO-based grafts. The results revealed that EO-based grafts showed favorable ability for bone regeneration. However, there was no significant difference in bone regeneration between EO-based grafts that utilized chondrogenic differentiation or hypertrophic differentiation. Additionally, the results demonstrated low quality in the experimental methods and the reporting of animal studies as well as a low quality of evidence provided by the included studies. Based on this, we propose three suggestions to enhance the quality of experimental methods and reporting in animal experiments. Furthermore, it is essential to conduct more evidence-based research to establish reliable evidence for the clinical application of EO-based grafts.
Collapse
Affiliation(s)
- Yihua Shi
- Department of Orthopedic Surgery, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 442000, China
| | - Ranning Zhuo
- Department of Clinical Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Weichun Guo
- Department of Orthopedic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yubo Shi
- Department of Orthopedic Surgery, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 442000, China
| |
Collapse
|
3
|
Zhou W, Liu Y, Nie X, Zhu C, Xiong L, Zhou J, Huang W. Peptide-based inflammation-responsive implant coating sequentially regulates bone regeneration to enhance interfacial osseointegration. Nat Commun 2025; 16:3283. [PMID: 40189598 PMCID: PMC11973180 DOI: 10.1038/s41467-025-58444-8] [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: 09/22/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Aseptic loosening is the primary cause of bone prosthesis failure, commonly attributed to inadequate osseointegration due to coatings misaligned with bone regeneration. Here, we modify the titanium surface with a mussel-inspired peptide to form a 3,4-dihydroxyphenylalanine (DOPA)-rich coating, then graft N3-K15-PVGLIG-K23 (P1) and N3-Y5-PVGLIG-K23 (P2), which are composed of anti-inflammatory (K23), angiogenic (K15), osteogenic (Y5), and inflammation-responsive (PVGLIG) sequences, onto the surface via click chemistry, forming the DOPA-P1@P2 coating. DOPA-P1@P2 promotes bone regeneration through sequential regulation. In the initial stage, the outermost K23 induces M2 macrophage polarization, establishing a pro-regenerative immune microenvironment. Subsequently, K15 and Y5, exposed by the release of K23, enhance angiogenesis and osteogenesis. In the final stage, DOPA-P1@P2 outperforms the TiO₂ control, showing a 161% increase in maximal push-out force, a 207% increase in bone volume fraction, and a 1409% increase in bone-to-implant contact. These findings show that DOPA-P1@P2 efficiently enhances interfacial osseointegration by sequentially regulating bone regeneration, providing viable insights into coating design.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Liu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xuan Nie
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China
| | - Chen Zhu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Liming Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jing Zhou
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
4
|
Birjandi AA, Sharpe P. Therapeutic potential of curcumin in regenerative dentistry. FRONTIERS IN DENTAL MEDICINE 2025; 6:1537478. [PMID: 40196309 PMCID: PMC11973315 DOI: 10.3389/fdmed.2025.1537478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/17/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Natural compounds have emerged as promising candidates in drug development due to their potent immunomodulatory anti-inflammatory, antibacterial, analgesic, and healing properties. They have shown significant therapeutic potential in clinical applications, such as mouth rinses, toothpastes, and localized delivery systems. The use of natural alternatives can contribute to tackling antimicrobial resistance. Among natural compounds, curcumin has gained particular attention, demonstrating robust anti-cancer, antibiotic, and anti-inflammatory activities in numerous in vivo studies, while exhibiting a favorable safety profile for the treatment of various diseases. In this study, the remedial effects of curcumin and its metabolite, tetrahydrocurcumin, on dental pulp were explored. In addition, these results were compared with our previous findings on the effects of these natural compounds on periodontal ligament and gingival epithelial cells, further broadening our understanding of their therapeutic potential in oral disease such as caries and periodontitis. Methods RNA sequencing was used to investigate the differentially expressed genes in dental pulp cells following treatments with curcumin and tetrahydrocurcumin. Results We show that treatment of dental pulp cells with 1 μM of curcumin or tetrahydrocurcumin is sufficient to promote Wnt signaling pathway in dental pulp cells. Curcumin treatment promotes the upregulation of cellular metabolism and enhances cellular response to stress. Our enrichment analysis shows that treatment with tetrahydrocurcumin modulates the extracellular matrix and angiogenesis. Conclusions The findings of this study highlight the cytoprotective and regenerative properties of curcumin and tetrahydrocurcumin. These properties could be leveraged as a therapeutic approach to promote tissue regeneration in oral diseases.
Collapse
Affiliation(s)
| | - Paul Sharpe
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, Kings College London, London, United Kingdom
| |
Collapse
|
5
|
Zhou L, Cai W, Zhang Y, Zhong W, He P, Ren J, Gao X. Therapeutic effect of mesenchymal stem cell-derived exosome therapy for periodontal regeneration: a systematic review and meta-analysis of preclinical trials. J Orthop Surg Res 2025; 20:27. [PMID: 39780243 PMCID: PMC11715287 DOI: 10.1186/s13018-024-05403-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND To assess the therapeutic effects of mesenchymal stem cell (MSC)-derived exosome therapy on periodontal regeneration and identify treatment factors associated with enhanced periodontal regeneration in recent preclinical studies. METHODS Searches were conducted in PubMed, Cochrane Library, EMBASE, and Web of Science databases until October 10, 2024. A risk of bias (ROB) assessment was performed using the SYRCLE tool. Osteogenic-related parameters were used as the primary outcome measures. RESULTS In total, 1360 articles were identified, of which 17 preclinical studies were based on MSC-derived exosome therapy, and they demonstrated a beneficial effect on BV/TV (SMD = 13.99; 95% Cl = 10.50, 17.48; p < 0.00001), CEJ-ABC (SMD = -0.22; 95% Cl = -0.31, -0.13; p < 0.00001), BMD (SMD = 0.29; 95% Cl = 0.14, 0.45; p = 0.0002), and Tp.Sp (SMD = -0.08; 95% Cl= -0.15, -0.02; p = 0.02) compared with the control group. However, no significant differences were observed in Tp.Th (SMD = 0.03; 95% CI = 0.00, 0.07; p = 0.09) between the exosome-treated group and control group. Additionally, subgroup analysis indicated that preconditioned exosomes (p = 0.03) significantly improved BV/TV. In contrast, there were no significant differences in the enhancement of BV/TV with respect to the application method (p = 0.29), application frequency (p = 0.10), treatment duration (p = 0.15), or source of MSCs (p = 0.31). CONCLUSIONS MSC-derived exosomes show great promise for enhancing the quality of periodontal regeneration. However, more standardized and robust trials are needed to reduce heterogeneity and bias across studies and to confirm the therapeutic parameters associated with the enhancement of periodontal regeneration by MSC-derived exosomes. REGISTRATION CRD42024546236.
Collapse
Affiliation(s)
- Liping Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Wenjia Cai
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Yuhan Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Wenjie Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Ping He
- Department of Stomatology, Dazhou Central Hospital, Dazhou, 635000, China.
| | - Jingsong Ren
- Department of Stomatology, Dazhou Central Hospital, Dazhou, 635000, China.
| | - Xiang Gao
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China.
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China.
| |
Collapse
|
6
|
Heinämäki J, Koshovyi O, Botsula I, Shpychak A, Vo HQ, Nguyen HT, Raal A. Plant-Origin Compounds and Materials for Advancing Bone Tissue Engineering and 3D Bioprinting: Traditional Medicine Aspects and Current Perspectives. J Tissue Eng Regen Med 2025; 2025:2812191. [PMID: 40224956 PMCID: PMC11985229 DOI: 10.1155/term/2812191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/13/2024] [Indexed: 04/15/2025]
Abstract
Bone defects are becoming a true challenge in global health care due to the aging population and higher prevalence of musculoskeletal disorders. The interest in using plant-origin compounds and plant-derived biomaterials in bone tissue engineering (BTE) has been increased due to their availability (abundance), safety, biocompatibility, biodegradability, and low cost. Plant-origin compounds have supportive effects on bone tissue healing, and cell-laden plant-derived biomaterials can be applied in formulating bioinks for three-dimensional (3D) bioprinting to facilitate the preparation of native bone tissue-mimicking structures and customized bone scaffolds. Such plant-derived materials also have the capacity to improve cell viability and support osteoconductive and osteoinductive properties of a bone construct. In this article, we review the ethnomedical aspects related to the use of medicinal plants and plant-origin bioactive compounds in bone healing and the recent developments in the 3D bioprinting of bone constructs with plant-derived biomaterials for advancing BTE. The commonly used 3D-bioprinting techniques, the properties of plant-origin compounds and biomaterials (for bone 3D bioprinting), and the selective examples of bone scaffolds fabricated using plant-derived biomaterials are discussed with a special reference set on applicability, performance, advantages, limitations, and challenges. Plant-origin compounds, biomaterials, and biomimetic 3D-bioprinted constructs could be the basis for a next-generation BTE.
Collapse
Affiliation(s)
- Jyrki Heinämäki
- Institute of Pharmacy, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Oleh Koshovyi
- Institute of Pharmacy, Faculty of Medicine, University of Tartu, Tartu, Estonia
- Department of Pharmacognosy, National University of Pharmacy, Kharkiv, Ukraine
| | - Iryna Botsula
- Department of Pharmacognosy, National University of Pharmacy, Kharkiv, Ukraine
| | - Alina Shpychak
- Department of Pharmacognosy, National University of Pharmacy, Kharkiv, Ukraine
| | - Hung Quoc Vo
- Faculty of Pharmacy, University of Medicine and Pharmacy, Hue University, Hue, Vietnam
| | - Hoai Thi Nguyen
- Faculty of Pharmacy, University of Medicine and Pharmacy, Hue University, Hue, Vietnam
| | - Ain Raal
- Institute of Pharmacy, Faculty of Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
7
|
Liu Y, Jiang G, Sun M, Zhou Z, Liang P, Chang Q. DeepTransformer: Node Classification Research of a Deep Graph Network on an Osteoporosis Graph based on GraphTransformer. Curr Comput Aided Drug Des 2025; 21:28-37. [PMID: 39651564 DOI: 10.2174/0115734099266731231115065030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 12/11/2024]
Abstract
BACKGROUND Osteoporosis (OP) is one of the most common diseases in the elderly population. It is mostly treated with medication, but drug research and development have the disadvantage of taking a long time and having a high cost. OBJECTIVE Therefore, we developed a graph neural network with the help of artificial intelligence to provide new ideas for drug research and development for OP. METHODS In this study, we built a new osteoporosis graph (called OPGraph) and proposed a deep graph neural network (called DeepTransformer) to predict new drugs for OP. OPGraph is a graph data model established by gathering features and their interrelationships from a vast amount of OP data. DeepTransformer uses GraphTransformer as its foundational network and applies residual connections for deep layering. RESULTS The analysis and results showed that DeepTransformer outperformed numerous models on OPGraph, with area under the curve (AUC) and area under the precision-recall curve (AUPR) reaching 0.9916 and 0.9911, respectively. In addition, we conducted an in vitro validation experiment on two of the seven predicted compounds (Puerarin and Aucubin), and the results corroborated the predictions of our model. CONCLUSION The model we developed with the help of artificial intelligence can effectively reduce the time and cost of OP drug development and reduce the heavy economic burden brought to patient's family by complications caused by osteoporosis.
Collapse
Affiliation(s)
- Yixin Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Guowei Jiang
- Pharmacy Department, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Miaomiao Sun
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Ziyan Zhou
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Pengchen Liang
- School of Microelectronics, Shanghai University, Shanghai, 201800, China
| | - Qing Chang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China
| |
Collapse
|
8
|
Hu Z, Lin H, Wang Z, Yi Y, Zou S, Liu H, Han X, Rong X. 3D Printing Hierarchical Porous Nanofibrous Scaffold for Bone Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405406. [PMID: 39548932 DOI: 10.1002/smll.202405406] [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: 07/01/2024] [Revised: 09/16/2024] [Indexed: 11/18/2024]
Abstract
Current limitations in 3D printing pose significant challenges for the fabrication of hierarchical 3D scaffolds with nanofibrous structures that simulate the natural bone extracellular matrix (ECM) for enhanced bone regeneration. This study presents an innovative approach to 3D printing customized hierarchical porous scaffolds with nanofiber structures using biodegradable nanofibrous microspheres as the bio-ink. In vitro investigations demonstrate that the hierarchical porous architecture substantially enhances cell infiltration and proliferation rates, while the nanofiber topology provides physical cues to guide osteogenic differentiation and ECM deposition. When serving as a cell carrier, the 3D-printed nanofibrous scaffold promotes bone tissue regeneration and integration in vivo. Additionally, the facile and versatile chemical modification facilitates the precise tailoring of the scaffold's functionality. Using nanofibrous microspheres with highly biomimetic and versatile modification properties as the foundational constituent in this universal 3D printing methodology enables comprehensive manipulation of scaffolding biological properties, spanning from macroscopic external morphology to molecular-scale biochemical kinetics, thereby addressing a diverse spectrum of clinical requisites.
Collapse
Affiliation(s)
- Zhiai Hu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hengyi Lin
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhenming Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yating Yi
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hao Liu
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xin Rong
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
9
|
Khalkhali P, Omidi M, Masson-Meyers DS, Akbari B, Dehghan MM, Aminianfar H, Farzad-Mohajeri S, Mansouri V, Nikpasand A, Tayebi L. Promoting Angiogenesis/Osteogenesis by a New Copper/Magnesium Hydroxide Hybrid Nanoparticle: In Vitro and In Vivo Investigation. J Biomed Mater Res A 2025; 113:e37855. [PMID: 39815692 PMCID: PMC12068790 DOI: 10.1002/jbm.a.37855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
In this study, a new hybrid nanoparticle composed of magnesium hydroxide and copper oxide (Mg(OH)2/CuO) with an optimized ratio of magnesium (Mg) to copper (Cu) was designed and incorporated into a 3D-printed scaffold made of polycaprolactone (PCL) and gelatin. These hybrid nanostructures (MCNs) were prepared using a green, solvent-free method. Their topography, surface morphology, and structural properties were characterized using scanning electron microscopy (SEM), transmission electron microscopy (TEM), X-ray diffraction (XRD), and X-ray photoelectron spectroscopy (XPS). The fabricated 3D-printed PCL/Gelatin/MCN scaffolds were investigated in vitro and in vivo. Cell viability tests on murine calvarial preosteoblasts (MC3T3-E1) and human umbilical vein endothelial cells (HUVECs) demonstrated that the scaffolds could induce proper cell proliferation. Additionally, the angiogenic and osteogenic properties of the constructs were evaluated using alkaline phosphatase (ALP) activity, osteogenesis-related, and angiogenesis-related gene expression tests. The in vivo study was conducted using a rat calvarial defect model, which confirmed the superior angiogenic and osteogenic properties of the PCL/gelatin/MCN scaffolds compared to PCL/Gelatin and PCL/Gelatin/Mg(OH)2 scaffolds. Overall, the PCL/Gelatin/MCN scaffolds showed promising potential for bone regeneration, particularly for critical-sized defects where proper angiogenesis is essential for tissue reconstruction.
Collapse
Affiliation(s)
- Parsa Khalkhali
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Meisam Omidi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | | | - Babak Akbari
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Veterinary Clinical Sciences, Karaj Branch, Islamic Azad University, Karaj, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Hossein Aminianfar
- Department of Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Saeed Farzad-Mohajeri
- Department of Veterinary Clinical Sciences, Karaj Branch, Islamic Azad University, Karaj, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Vahid Mansouri
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical sciences, Tehran 19857-17443, Iran
- Department of Basic Science, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | - Amin Nikpasand
- Department of Veterinary Clinical Sciences, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
- Institute for Engineering in Medicine, Health, & Human Performance (EnMed), Batten College of Engineering and Technology, Old Dominion University, Norfolk, VA, 23529, USA
| |
Collapse
|
10
|
Bose S, Chaudhari VS, Kushram P. 3D printed scaffolds with quercetin and vitamin D3 nanocarriers: In vitro cellular evaluation. J Biomed Mater Res A 2024; 112:2110-2123. [PMID: 38894584 PMCID: PMC11464199 DOI: 10.1002/jbm.a.37756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024]
Abstract
Increasing bone diseases and anomalies significantly challenge bone regeneration, necessitating the development of innovative implantable devices for effective healing. This study explores the potential of 3D-printed calcium phosphate (CaP) scaffolds functionalized with natural medicine to address this issue. Specifically, quercetin and vitamin D3 (QVD) encapsulated solid lipid nanoparticles (QVD-SLNs) are incorporated into the scaffold to enhance bone regeneration. The melt emulsification method is utilized to achieve high drug encapsulation efficiency (~98%) and controlled biphasic release kinetics. The process-structure-property performance of these systems allows more controlled release while maintaining healthy cell-material interactions. The functionalized scaffolds show ~1.3- and ~-1.6-fold increase in osteoblast cell proliferation and differentiation, respectively, as compared with the control. The treated scaffold demonstrates a reduction in osteoclastic activity as compared with the control. The QVD-SLN-loaded scaffolds show ~4.2-fold in vitro chemopreventive potential against osteosarcoma cells. Bacterial assessment with both Staphylococcus aureus and Pseudomonas aeruginosa shows a significant reduction in bacterial colony growth over the treated scaffold. These findings summarize that the release of QVD-SLNs through a 3D-printed CaP scaffold can treat various bone-related disorders for low or non-load-bearing applications.
Collapse
Affiliation(s)
- Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington, USA
| | - Vishal Sharad Chaudhari
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington, USA
| | - Priya Kushram
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington, USA
| |
Collapse
|
11
|
Li X, Sun Z, Shang X, Chen L, Shi X, Xu W, Fu S, He Q, Liang Q, Ma J, Sun X, Lu J, Jin W. Sequential delivery of IL-10 and icariin using nanoparticle/hydrogel hybrid system for prompting bone defect repair. Mater Today Bio 2024; 29:101374. [PMID: 39717024 PMCID: PMC11664418 DOI: 10.1016/j.mtbio.2024.101374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/11/2024] [Accepted: 11/28/2024] [Indexed: 12/25/2024] Open
Abstract
The treatment of large bone defects remains challenging due to the lack of spatiotemporal management of the immune microenvironment, inflammation response and bone remodeling. To address these issues, we designed and developed a nanoparticle/hydrogel hybrid system that can achieve the combined and sequential delivery of an anti-inflammatory factor (IL-10) and osteogenic drug (icariin, ICA). A photopolymerizable composite hydrogel was prepared by combining gelatin methacryloyl (GelMA) and heparin-based acrylated hyaluronic acid (HA) hydrogels containing IL-10, and poly(dl-lactide-co-glycolide) (PLGA)-HA nanoparticles loaded with ICA were incorporated into the composite hydrogels. The nanoparticle/hydrogel hybrid system demonstrates an array of features including mechanical strength, injectability and photo-crosslinking. The rapid release of IL-10 from the hydrogel effectively exerts immunomodulatory activity, whereas the long-term sustained release of icariin from the PLGA-HA nanoparticles significantly triggers the osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Notably, the combined delivery of IL-10 and ICA from the hybrid system exhibit a synergistic effect for bone remodeling in a critical cranial defect rat model. Our findings indicate the importance of the immunomodulatory microenvironment and osteogenic differentiation for high-quality skull remodeling, and thus the dual-factor releasing nanoparticle/hydrogel hybrid system could be a promising candidate for repairing bone defects.
Collapse
Affiliation(s)
- Xiaojun Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Zeyue Sun
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xiushuai Shang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Liuting Chen
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xiaofeng Shi
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Wei Xu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Shaotian Fu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Qingling He
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Qihao Liang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Jie Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Xin Sun
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Jiaju Lu
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Wenjie Jin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| |
Collapse
|
12
|
Dahiya A, Chaudhari VS, Bose S. Bone Healing via Carvacrol and Curcumin Nanoparticle on 3D Printed Scaffolds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2405642. [PMID: 39463050 PMCID: PMC11636189 DOI: 10.1002/smll.202405642] [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: 07/07/2024] [Revised: 09/22/2024] [Indexed: 10/29/2024]
Abstract
Carvacrol is a potent antimicrobial and anti-inflammatory agent, while curcumin possesses antioxidant, anti-inflammatory, and anticancer properties. These phytochemicals have poor solubility, bioavailability, and stability in their free form. Nanoencapsulation can reduce these limitations with enhanced translational capability. Integrating nanocarriers with 3D-printed calcium phosphate (CaP) scaffolds presents a novel strategy for bone regeneration. Carvacrol and curcumin-loaded nanoparticles (CC-NP) synthesized with melt emulsification produced negatively charged, monodispersed particles with a hydrodynamic diameter of ≈127 nm. Their release from the scaffold shows a biphasic release under physiological and acidic conditions. At pH 5.0, the CC-NP exhibits a 53% release of curcumin and nearly 100% release of carvacrol, compared to 19% and 36% from their respective drug solutions. At pH 7.4, ≈40% of curcumin and 76% of carvacrol releases, highlighting their pH-sensitive release mechanism. In vitro studies demonstrate a 1.4-fold increase in osteoblast cell viability with CC-NP treatment. CC-NP exhibit cytotoxic effects against osteosarcoma cells, reducing cell viability by ≈2.9-fold. The antibacterial efficacy of CC-NP evaluated against Staphylococcus aureus (SA) and Pseudomonas aeruginosa (PA) exhibiting 98% antibacterial efficacy. This approach enhances therapeutic outcomes and minimizes the potential side effects associated with conventional treatments, paving the way for innovative applications in regenerative medicine.
Collapse
Affiliation(s)
- Aditi Dahiya
- W. M. Keck Biomedical Materials Research LaboratorySchool of Mechanical and Materials EngineeringWashington State UniversityPullmanWashington99164USA
- Department of ChemistryWashington State UniversityPullmanWashington99164USA
| | - Vishal Sharad Chaudhari
- W. M. Keck Biomedical Materials Research LaboratorySchool of Mechanical and Materials EngineeringWashington State UniversityPullmanWashington99164USA
| | - Susmita Bose
- W. M. Keck Biomedical Materials Research LaboratorySchool of Mechanical and Materials EngineeringWashington State UniversityPullmanWashington99164USA
- Department of ChemistryWashington State UniversityPullmanWashington99164USA
| |
Collapse
|
13
|
Zhao X, Li L, Zhang Y, Liu Z, Xing H, Gu Z. 3D Printing and Property of Biomimetic Hydroxyapatite Scaffold. Biomimetics (Basel) 2024; 9:714. [PMID: 39590286 PMCID: PMC11591832 DOI: 10.3390/biomimetics9110714] [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: 10/12/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
The 3D printing of a biomimetic scaffold with a high hydroxyapatite (HA) content (>80%) and excellent mechanical property is a serious challenge because of the difficulty of forming and printing, insufficient cohesion, and low mechanical property of the scaffold. In this study, hydroxyapatite whiskers (HAWs), with their superior mechanical property, biodegradability, and biocompatibility, were used to reinforce spherical HA scaffolds by 3D printing. The compressive strength and energy absorption capacity of HAW-reinforced spherical HA (HAW/HA) scaffolds increased when the HAW/HA ratio increased from 0:10 to 4:6 and then dropped with any further increases in the HAW/HA ratio. Bioceramic content (HAWs and spherical HA) in the scaffolds reached 83%, and the scaffold with a HAW/HA ratio of 4:6 (4-HAW/HA) exhibited an optimum compressive strength and energy absorption capacity. The scaffold using polyvinyl alcohol (PVA) as an additive possessed a good bonding between HA and PVA as well as a higher strength, which allowed the scaffold to bear a higher stress at the same strain. The compressive strength and toughness of the 4-HAW/HA-PVA scaffold were 1.96 and 1.63 times that of the 4-HAW/HA scaffold with hydroxypropyl methyl cellulose (HPMC), respectively. The mechanical property and inorganic components of the biomimetic HAW/HA scaffold were similar to those of human bone, which would make it ideal for repairing bone defects.
Collapse
Affiliation(s)
- Xueni Zhao
- College of Mechanical and Electrical Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (L.L.)
| | | | | | | | | | | |
Collapse
|
14
|
Xie Q, Wang T, He L, Liang H, Sun J, Huang X, Xie W, Niu Y. Biological and structural properties of curcumin-loaded graphene oxide incorporated collagen as composite scaffold for bone regeneration. Front Bioeng Biotechnol 2024; 12:1505102. [PMID: 39634102 PMCID: PMC11614606 DOI: 10.3389/fbioe.2024.1505102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction To address the challenges related to bone defects, including osteoinductivity deficiency and post-implantation infection risk, this study developed the collagen composite scaffolds (CUR-GO-COL) with multifunctionality by integrating the curcumin-loaded graphene oxide with collagen through a freeze-drying-cross-linking process. Methods The morphological and structural characteristics of the composite scaffolds were analyzed, along with their physicochemical properties, including water absorption capacity, water retention rate, porosity, in vitro degradation, and curcumin release. To evaluate the biocompatibility, cell viability, proliferation, and adhesion capabilities of the composite scaffolds, as well as their osteogenic and antimicrobial properties, in vitro cell and bacterial assays were conducted. These assays were designed to assess the impact of the composite scaffolds on cell behavior and bacterial growth, thereby providing insights into their potential for promoting osteogenesis and inhibiting infection. Results The CUR-GO-COL composite scaffold with a CUR-GO concentration of 0.05% (w/v) exhibits optimal biological compatibility and stable and slow curcumin release rate. Furthermore, in vitro cell and bacterial tests demonstrated that the prepared CUR-GO-COL composite scaffolds enhance cell viability, proliferation and adhesion, and offer superior osteogenic and antimicrobial properties compared with the CUR-GO composite scaffold, confirming the osteogenesis promotion and antimicrobial effects. Discussion The introduction of CUR-GO into collagen scaffold creates a bone-friendly microenvironment, and offers a theoretical foundation for the design, investigation and utilization of multifunctional bone tissue biomaterials.
Collapse
Affiliation(s)
- Qi Xie
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Tianqi Wang
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Lina He
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Hongbo Liang
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin, China
| | - Jingxuan Sun
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Xiaoxiao Huang
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin, China
| | - Weili Xie
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Yumei Niu
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| |
Collapse
|
15
|
Gupta S, Qayoom I, Mairal A, Singh S, Kumar A. Local Delivery of Exosomes and Antibiotics in Hydroxyapatite-Based Nanocement for Osteomyelitis Management. ACS Infect Dis 2024; 10:3994-4008. [PMID: 39469832 DOI: 10.1021/acsinfecdis.4c00721] [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: 10/30/2024]
Abstract
The management of bone and joint infections is a formidable challenge in orthopedics and poses a global health concern. While clinical management emphasizes infection prevention and complete eradication, an effective strategy for stabilizing skeletal tissue with adequate soft tissue coverage remains limited. In this study, we have employed a novel approach of using the local delivery of exosomes and antibiotics (rifampicin) using a hydroxyapatite-based nanocement carrier to manage the residual space created during debridement effectively. Additionally, we synthesized a periosteum-guiding antioxidant herbal membrane to leverage the inherent periosteum regeneration capability of the bone, facilitating bone callus repair and natural healing. The synthesized scaffolds were biocompatible and demonstrated potent antibacterial activity in vitro. When evaluated in vivo in the Staphylococcus aureus-induced rat tibial osteomyelitis model, the released drugs successfully cleared the residual bacteria and the released exosome promoted bone healing, resulting in 3-fold increase in bone volume as analyzed via micro-CT analysis. Immunofluorescence staining of periosteum-specific markers also indicated the complete formation of periosteal layers, thus highlighting the complete bone repair.
Collapse
Affiliation(s)
- Sneha Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| | - Irfan Qayoom
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| | - Ayushi Mairal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| | - Sneha Singh
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Centre of Excellence in Orthopaedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| |
Collapse
|
16
|
Yang S, Wu H, Peng C, He J, Pu Z, Lin Z, Wang J, Hu Y, Su Q, Zhou B, Yong X, Lan H, Hu N, Hu X. From the microspheres to scaffolds: advances in polymer microsphere scaffolds for bone regeneration applications. BIOMATERIALS TRANSLATIONAL 2024; 5:274-299. [PMID: 39734699 PMCID: PMC11681185 DOI: 10.12336/biomatertransl.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 12/31/2024]
Abstract
The treatment and repair of bone tissue damage and loss due to infection, tumours, and trauma are major challenges in clinical practice. Artificial bone scaffolds offer a safer, simpler, and more feasible alternative to bone transplantation, serving to fill bone defects and promote bone tissue regeneration. Ideally, these scaffolds should possess osteoconductive, osteoinductive, and osseointegrative properties. However, the current first-generation implants, represented by titanium alloys, have shown poor bone-implant integration performance and cannot meet the requirements for bone tissue repair. This has led to increased research on second and third generation artificial bone scaffolds, which focus on loading bioactive molecules and cells. Polymer microspheres, known for their high specific surface areas at the micro- and nanoscale, exhibit excellent cell and drug delivery behaviours. Additionally, with their unique rigid structure, microsphere scaffolds can be constructed using methods such as thermal sintering, injection, and microsphere encapsulation. These scaffolds not only ensure the excellent cell drug loading performance of microspheres but also exhibit spatial modulation behaviour, aiding in bone repair within a three-dimensional network structure. This article provides a summary and discussion of the use of polymer microsphere scaffolds for bone repair, focusing on the mechanisms of bone tissue repair and the current status of clinical bone grafts, aimed at advancing research in bone repair.
Collapse
Affiliation(s)
- Shuhao Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Haoming Wu
- School of Preclinical Medicine of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Chao Peng
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Jian He
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Zhengguang Pu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Zhidong Lin
- The Second Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Jun Wang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yingkun Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Qiao Su
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Bingnan Zhou
- School of Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Yong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan Province, China
| | - Hai Lan
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
17
|
Toulou C, Chaudhari VS, Bose S. Extrusion 3D-printed tricalcium phosphate-polycaprolactone biocomposites for quercetin-KCl delivery in bone tissue engineering. J Biomed Mater Res A 2024; 112:1472-1483. [PMID: 38477071 PMCID: PMC11239310 DOI: 10.1002/jbm.a.37692] [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: 10/02/2023] [Revised: 01/12/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024]
Abstract
Critical-sized bone defects pose a significant challenge in advanced healthcare due to limited bone tissue regenerative capacity. The complex interplay of numerous overlapping variables hinders the development of multifunctional biocomposites. Phytochemicals show promise in promoting bone growth, but their dose-dependent nature and physicochemical properties halt clinical use. To develop a comprehensive solution, a 3D-printed (3DP) extrusion-based tricalcium phosphate-polycaprolactone (TCP-PCL) scaffold is augmented with quercetin and potassium chloride (KCl). This composite material demonstrates a compressive strength of 30 MPa showing promising stability for low load-bearing applications. Quercetin release from the scaffold follows a biphasic pattern that persists for up to 28 days, driven via diffusion-mediated kinetics. The incorporation of KCl allows for tunable degradation rates of scaffolds and prevents the initial rapid release. Functionalization of scaffolds facilitates the attachment and proliferation of human fetal osteoblasts (hfOB), resulting in a 2.1-fold increase in cell viability. Treated scaffolds exhibit a 3-fold reduction in osteosarcoma (MG-63) cell viability as compared to untreated substrates. Ruptured cell morphology and decreased mitochondrial membrane potential indicate the antitumorigenic potential. Scaffolds loaded with quercetin and quercetin-KCl (Q-KCl) demonstrate 76% and 89% reduction in bacterial colonies of Staphylococcus aureus, respectively. This study provides valuable insights as a promising strategy for bone tissue engineering (BTE) in orthopedic repair.
Collapse
Affiliation(s)
- Connor Toulou
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington, USA
| | - Vishal Sharad Chaudhari
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington, USA
| | - Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington, USA
| |
Collapse
|
18
|
Chen L, Xie Y, Chen X, Li H, Lu Y, Yu H, Zheng D. O-carboxymethyl chitosan in biomedicine: A review. Int J Biol Macromol 2024; 275:133465. [PMID: 38945322 DOI: 10.1016/j.ijbiomac.2024.133465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/01/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
O-carboxymethyl chitosan (O-CMC) is a chitosan derivative produced through the substitution of hydroxyl (-OH) functional groups in glucosamine units with carboxymethyl (-CH2COOH) substituents, effectively addressing the inherent solubility issues of chitosan in aqueous solutions. O-CMC has garnered significant interest due to its enhanced solubility, elevated viscosity, minimal toxicity, and advantageous biocompatibility properties. Furthermore, O-CMC demonstrates antibacterial, antifungal, and antioxidant characteristics, rendering it a promising candidate for various biomedical uses such as wound healing, tissue engineering, anti-tumor therapies, biosensors, and bioimaging. Additionally, O-CMC is well-suited for the fabrication of nanoparticles, hydrogels, films, microcapsules, and tablets, offering opportunities for effective drug delivery systems. This review outlines the distinctive features of O-CMC, offers analyses of advancements and future potential based on current research, examines significant obstacles for clinical implementation, and foresees its ongoing significant impacts in the realm of biomedicine.
Collapse
Affiliation(s)
- Lingbin Chen
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yandi Xie
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China; Department of Prosthodontics & Research Center of Dental Esthetics and Biomechanics, Fujian Medical University, Fuzhou, China
| | - Xiaohang Chen
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China; Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Hengyi Li
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Youguang Lu
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China; Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Hao Yu
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China; Department of Prosthodontics & Research Center of Dental Esthetics and Biomechanics, Fujian Medical University, Fuzhou, China.
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
19
|
Astaneh ME, Noori F, Fereydouni N. Curcumin-loaded scaffolds in bone regeneration. Heliyon 2024; 10:e32566. [PMID: 38961905 PMCID: PMC11219509 DOI: 10.1016/j.heliyon.2024.e32566] [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: 12/27/2023] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
In recent years, there has been a notable surge in the development of engineered bone scaffolds intended for the repair of bone defects. While autografts and allografts have traditionally served as the primary methods in bone tissue engineering, their inherent limitations have spurred the exploration of novel avenues in biomedical implant development. The emergence of bone scaffolds not only facilitates bone reconstruction but also offers a platform for the targeted delivery of therapeutic agents. There exists a pervasive interest in leveraging various drugs, proteins, growth factors, and biomolecules with osteogenic properties to augment bone formation, as the enduring side effects associated with current clinical modalities necessitate the pursuit of safer alternatives. Curcumin, the principal bioactive compound found in turmeric, has demonstrated notable efficacy in regulating the proliferation and differentiation of bone cells while promoting bone formation. Nevertheless, its utility is hindered by restricted water solubility and poor bioavailability. Strategies aimed at enhancing the solubility, stability, and bioavailability of curcumin, including formulation techniques such as liposomes and nanoparticles or its complexation with metals, have been explored. This investigation is dedicated to exploring the impact of curcumin on the proliferation, differentiation, and migration of osteocytes, osteoblasts, and osteoclasts.
Collapse
Affiliation(s)
- Mohammad Ebrahim Astaneh
- Department of Anatomical Sciences, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Fariba Noori
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Narges Fereydouni
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
20
|
Wan Z, Bai X, Wang X, Guo X, Wang X, Zhai M, Fu Y, Liu Y, Zhang P, Zhang X, Yang R, Liu Y, Lv L, Zhou Y. Mgp High-Expressing MSCs Orchestrate the Osteoimmune Microenvironment of Collagen/Nanohydroxyapatite-Mediated Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308986. [PMID: 38588510 PMCID: PMC11187922 DOI: 10.1002/advs.202308986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/22/2024] [Indexed: 04/10/2024]
Abstract
Activating autologous stem cells after the implantation of biomaterials is an important process to initiate bone regeneration. Although several studies have demonstrated the mechanism of biomaterial-mediated bone regeneration, a comprehensive single-cell level transcriptomic map revealing the influence of biomaterials on regulating the temporal and spatial expression patterns of mesenchymal stem cells (MSCs) is still lacking. Herein, the osteoimmune microenvironment is depicted around the classical collagen/nanohydroxyapatite-based bone repair materials via combining analysis of single-cell RNA sequencing and spatial transcriptomics. A group of functional MSCs with high expression of matrix Gla protein (Mgp) is identified, which may serve as a pioneer subpopulation involved in bone repair. Remarkably, these Mgp high-expressing MSCs (MgphiMSCs) exhibit efficient osteogenic differentiation potential and orchestrate the osteoimmune microenvironment around implanted biomaterials, rewiring the polarization and osteoclastic differentiation of macrophages through the Mdk/Lrp1 ligand-receptor pair. The inhibition of Mdk/Lrp1 activates the pro-inflammatory programs of macrophages and osteoclastogenesis. Meanwhile, multiple immune-cell subsets also exhibit close crosstalk between MgphiMSCs via the secreted phosphoprotein 1 (SPP1) signaling pathway. These cellular profiles and interactions characterized in this study can broaden the understanding of the functional MSC subpopulations at the early stage of biomaterial-mediated bone regeneration and provide the basis for materials-designed strategies that target osteoimmune modulation.
Collapse
Affiliation(s)
- Zhuqing Wan
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xiaoqiang Bai
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xin Wang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xiaodong Guo
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xu Wang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Mo Zhai
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Yang Fu
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Yunsong Liu
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Ping Zhang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xiao Zhang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Ruili Yang
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
- Department of OrthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
| | - Yan Liu
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
- Department of OrthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
| | - Longwei Lv
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Yongsheng Zhou
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| |
Collapse
|
21
|
Alsmael MA, Al-Khafaji AM. Evaluation of High-Performance Polyether Ether Ketone Polymer Treated with Piranha Solution and Epigallocatechin-3-Gallate Coating. BIOMED RESEARCH INTERNATIONAL 2024; 2024:1741539. [PMID: 38628498 PMCID: PMC11019569 DOI: 10.1155/2024/1741539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/27/2024] [Accepted: 03/23/2024] [Indexed: 04/19/2024]
Abstract
Background Dental implantation has become a standard procedure with high success rates, relying on achieving osseointegration between the implant surface and surrounding bone tissue. Polyether ether ketone (PEEK) is a promising alternative to traditional dental implant materials like titanium, but its osseointegration capabilities are limited due to its hydrophobic nature and reduced surface roughness. Objective The aim of the study is to increase the surface roughness and hydrophilicity of PEEK by treating the surface with piranha solution and then coating the surface with epigallocatechin-3-gallate (EGCG) by electrospraying technique. Materials and Methods The study includes four groups intended to investigate the effect of piranha treatment and EGCG coating: a control group of PEEK discs with no treatment (C), PEEK samples treated with piranha solution (P), a group of PEEK samples coated with EGCG (E), and a group of PEEK samples treated with piranha solution and coated with EGCG (PE). Surface roughness, wettability, and microhardness were assessed through statistical analysis. Results Piranha treatment increased surface roughness, while EGCG coating moderated it, resulting in an intermediate roughness in the PE group. EGCG significantly improved wettability, as indicated by the reduced contact angle. Microhardness increased by about 20% in EGCG-coated groups compared to noncoated groups. Statistical analysis confirmed significant differences between groups in all tests. Conclusion This study demonstrates the potential of EGCG coating to enhance the surface properties of PEEK as dental implants. The combined piranha and EGCG modification approach shows promise for improved osseointegration, although further vivo research is necessary. Surface modification techniques hold the key to optimizing biomaterial performance, bridging the gap between laboratory findings and clinical implementation in dental implantology.
Collapse
Affiliation(s)
- Mohammed A. Alsmael
- Department of Prosthodontics, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | | |
Collapse
|
22
|
Liu X, Gao J, Liu J, Cheng J, Han Z, Li Z, Chang Z, Zhang L, Li M, Tang P. Three-Dimensional-Printed Spherical Hollow Structural Scaffolds for Guiding Critical-Sized Bone Regeneration. ACS Biomater Sci Eng 2024; 10:2581-2594. [PMID: 38489227 DOI: 10.1021/acsbiomaterials.3c01956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
The treatment of bone tissue defects continues to be a complex medical issue. Recently, three-dimensional (3D)-printed scaffold technology for bone tissue engineering (BTE) has emerged as an important therapeutic approach for bone defect repair. Despite the potential of BTE scaffolds to contribute to long-term bone reconstruction, there are certain challenges associated with it including the impediment of bone growth within the scaffolds and vascular infiltration. These difficulties can be resolved by using scaffold structural modification strategies that can effectively guide bone regeneration. This study involved the preparation of biphasic calcium phosphate spherical hollow structural scaffolds (SHSS) with varying pore sizes using 3D printing (photopolymerized via digital light processing). The chemical compositions, microscopic morphologies, mechanical properties, biocompatibilities, osteogenic properties, and impact on repairing critical-sized bone defects of SHSS were assessed through characterization analyses, in vitro cytological assays, and in vivo biological experiments. The results revealed the biomimetic properties of SHSS and their favorable biocompatibility. The scaffolds stimulated cell adhesion, proliferation, differentiation, and migration and facilitated the expression of osteogenic genes and proteins, including Col-1, OCN, and OPN. Furthermore, they could effectively repair a critical-sized bone defect in a rabbit femoral condyle by establishing an osteogenic platform and guiding bone regeneration in the defect region. This innovative strategy presents a novel therapeutic approach for assessing critical-sized bone defects.
Collapse
Affiliation(s)
- Xiao Liu
- Medical School of Chinese PLA, Beijing 100853, China
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Jianpeng Gao
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Jianheng Liu
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Junyao Cheng
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Zhenchuan Han
- Medical School of Chinese PLA, Beijing 100853, China
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Zijian Li
- Medical School of Chinese PLA, Beijing 100853, China
| | | | - Licheng Zhang
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Ming Li
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Peifu Tang
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| |
Collapse
|
23
|
Yang A, Wang Y, Feng Q, Fatima K, Zhang Q, Zhou X, He C. Integrating Fluorescence and Magnetic Resonance Imaging in Biocompatible Scaffold for Real-Time Bone Repair Monitoring and Assessment. Adv Healthc Mater 2024; 13:e2302687. [PMID: 37940192 DOI: 10.1002/adhm.202302687] [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: 08/16/2023] [Revised: 11/05/2023] [Indexed: 11/10/2023]
Abstract
In situ monitoring of bone tissue regeneration progression is critical for the development of bone tissue engineering scaffold. However, engineered scaffolds that can stimulate osteogenic progress and allow for non-invasive monitoring of in vivo bone regeneration simultaneously are rarely reported. Based on a hard-and-soft integration strategy, a multifunctional scaffold composed of 3D printed microfilaments and a hydrogel network containing simvastatin (SV), indocyanine green-loaded superamphiphiles, and aminated ultrasmall superparamagnetic iron oxide nanoparticles (USPIO-NH2 ) is fabricated. Both in vitro and in vivo results demonstrate that the as-prepared scaffold significantly promotes osteogenesis through controlled SV release. The biocomposite scaffold exhibits alkaline phosphatase-responsive near-infrared II fluorescence imaging. Meanwhile, USPIO-NH2 within the co-crosslinked nanocomposite network enables the visualization of scaffold degradation by magnetic resonance imaging. Therefore, the biocomposite scaffold enables or facilitates non-invasive in situ monitoring of neo-bone formation and scaffold degradation processes following osteogenic stimulation, offering a promising strategy to develop theranostic scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Ai Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yue Wang
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai, 201600, China
| | - Qian Feng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Kanwal Fatima
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Qianqian Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
24
|
Dahiya A, Chaudhari VS, Kushram P, Bose S. 3D Printed SiO 2-Tricalcium Phosphate Scaffolds Loaded with Carvacrol Nanoparticles for Bone Tissue Engineering Application. J Med Chem 2024; 67:2745-2757. [PMID: 38146876 PMCID: PMC11164277 DOI: 10.1021/acs.jmedchem.3c01884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Bone damage resulting from trauma or aging poses challenges in clinical settings that need to be addressed using bone tissue engineering (BTE). Carvacrol (CA) possesses anti-inflammatory, anticancer, and antibacterial properties. Limited solubility and physicochemical stability restrict its biological activity, requiring a stable carrier system for delivery. Here, we investigate the utilization of a three-dimensional printed (3DP) SiO2-doped tricalcium phosphate (TCP) scaffold functionalized with carvacrol-loaded lipid nanoparticles (CA-LNPs) to improve bone health. It exhibits a negative surface charge with an entrapment efficiency of ∼97% and size ∼129 nm with polydispersity index (PDI) and zeta potential values of 0.18 and -16 mV, respectively. CA-LNPs exhibit higher and long-term release over 35 days. The CA-LNP loaded SiO2-doped TCP scaffold demonstrates improved antibacterial properties against Staphylococcus aureus and Pseudomonas aeruginosa by >90% reduction in bacterial growth. Functionalized scaffolds result in 3-fold decrease and 2-fold increase in osteosarcoma and osteoblast cell viability, respectively. These findings highlight the therapeutic potential of the CA-LNP loaded SiO2-doped TCP scaffold for bone defect treatment.
Collapse
Affiliation(s)
- Aditi Dahiya
- Department of Chemistry, Washington State University, Pullman, Washington 99164, United States
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Vishal Sharad Chaudhari
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Priya Kushram
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Susmita Bose
- Department of Chemistry, Washington State University, Pullman, Washington 99164, United States
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| |
Collapse
|
25
|
Dong H, Cao Y, Zou K, Shao Q, Liu R, Zhang Y, Pan L, Ning B. Ellagic acid promotes osteoblasts differentiation via activating SMAD2/3 pathway and alleviates bone mass loss in OVX mice. Chem Biol Interact 2024; 388:110852. [PMID: 38145796 DOI: 10.1016/j.cbi.2023.110852] [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: 08/27/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
Characterized by bone mass loss, osteoporosis is an orthopedic disease typically found in postmenopausal women and aging individuals. Consistent with its pathogenesis summarized as an imbalance in bone formation/resorption, current pharmacologically therapeutic strategies for osteoporosis mainly aim to promote bone formation or/and inhibit bone resorption. However, few effective drugs with mild clinical side effects have been developed, making it a well-concerned issue to seek appropriate drugs for osteoporosis. In this study, we investigated the effect of ellagic acid (EA) on osteogenesis in vitro and in vivo and searched for its molecular mechanism. Here, we showed that EA promoted osteogenic differentiation of MSCs, increased mRNA and protein expression levels of osteoblast marker genes Runt-related transcription factor2, Osterix, Alkaline phosphatase, Collagen type I alpha 1, Osteopontin and Osteocalcin. Furthermore, ovariectomized mice with orally administered EA (10 mg/kg, 50 mg/kg) had significantly higher bone mass than those in controls. And experiments such as fluorescence double-labeling and enzyme-linked immunosorbent assay also demonstrated that EA could promote osteogenesis in vivo. To probe the molecular mechanism of EA, we performed RNA sequencing analysis using EA-treated BMSCs. Significant up-regulation of SMAD2/3 transcription factors was identified by RNA-seq, and it was confirmed in vitro that EA promoted bone formation by activating the SMAD2/3 signaling pathway. Evidence from our present experiments indicates that EA may be a promising candidate for clinical treatment for osteoporosis in future.
Collapse
Affiliation(s)
- Hui Dong
- Jinan Central Hospital, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China
| | - Yuxia Cao
- Jinan Central Hospital, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China
| | - Ke Zou
- Jinan Central Hospital, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China
| | - Qiang Shao
- Jinan Central Hospital, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China
| | - Ronghan Liu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China
| | - Liuzhu Pan
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China
| | - Bin Ning
- Jinan Central Hospital, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China.
| |
Collapse
|
26
|
Bose S, Sarkar N, Jo Y. Natural medicine delivery from 3D printed bone substitutes. J Control Release 2024; 365:848-875. [PMID: 37734674 PMCID: PMC11147672 DOI: 10.1016/j.jconrel.2023.09.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023]
Abstract
Unmet medical needs in treating critical-size bone defects have led to the development of numerous innovative bone tissue engineering implants. Although additive manufacturing allows flexible patient-specific treatments by modifying topological properties with various materials, the development of ideal bone implants that aid new tissue regeneration and reduce post-implantation bone disorders has been limited. Natural biomolecules are gaining the attention of the health industry due to their excellent safety profiles, providing equivalent or superior performances when compared to more expensive growth factors and synthetic drugs. Supplementing additive manufacturing with natural biomolecules enables the design of novel multifunctional bone implants that provide controlled biochemical delivery for bone tissue engineering applications. Controlled release of naturally derived biomolecules from a three-dimensional (3D) printed implant may improve implant-host tissue integration, new bone formation, bone healing, and blood vessel growth. The present review introduces us to the current progress and limitations of 3D printed bone implants with drug delivery capabilities, followed by an in-depth discussion on cutting-edge technologies for incorporating natural medicinal compounds embedded within the 3D printed scaffolds or on implant surfaces, highlighting their applications in several pre- and post-implantation bone-related disorders.
Collapse
Affiliation(s)
- Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States.
| | - Naboneeta Sarkar
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| | - Yongdeok Jo
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| |
Collapse
|
27
|
Zheng C, Zhang M. 3D-printed PCL/β-TCP/CS composite artificial bone and histocompatibility study. J Orthop Surg Res 2023; 18:981. [PMID: 38129861 PMCID: PMC10734195 DOI: 10.1186/s13018-023-04489-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 12/17/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Tissue-engineered bone materials are an effective tool to repair bone defects. In this study, a novel biodegradable polycaprolactone (PCL)/β-tricalcium phosphate (β-TCP)/calcium sulfate (CS) composite scaffold was prepared by using three-dimensional (3D) printing technology. METHODS Scanning electron microscopy, gas expansion displacement, and contact goniometry were used to examine the 3D-printed PCL/β-TCP/CS composite scaffolds. The results showed that the PCL/β-TCP/CS scaffolds possessed controllable porosity, hydrophobicity, biodegradability, and suitable apatite mineralization ability. To confirm the bone regenerative properties of the fabricated composite scaffolds, scaffold extracts were prepared and evaluated for their cytotoxicity to bone marrow mesenchymal stem cells (BMSCs) and their ability to induce and osteogenic differentiation in BMSCs. RESULTS The PCL/β-TCP/CS composite scaffolds induced a higher level of differentiation of BMSCs than the PCL scaffolds, which occurred through the expression of bone metastasis-related genes. The New Zealand white rabbit radial defect experiment further demonstrated that PCL/β-TCP/CS scaffolds could promote bone regeneration. CONCLUSIONS In summary, the 3D-printed PCL/β-TCP/CS composite porous artificial bone has good cytocompatibility, osteoinductivity, and histocompatibility, which make it an ideal bone material for tissue engineering.
Collapse
Affiliation(s)
- Chao Zheng
- Department of Orthopaedics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Mingman Zhang
- Department of Pediatric Liver Transplantation, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Rd., Chongqing, 400014, People's Republic of China.
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
28
|
Zhu Z, Lin Y, Li L, Liu K, Wen W, Ding S, Liu M, Lu L, Zhou C, Luo B. 3D Printing Drug-Free Scaffold with Triple-Effect Combination Induced by Copper-Doped Layered Double Hydroxides for the Treatment of Bone Defects. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58196-58211. [PMID: 38079497 DOI: 10.1021/acsami.3c13336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Tissue-engineered poly(l-lactide) (PLLA) scaffolds have been widely used to treat bone defects; however, poor biological activities have always been key challenges for its further application. To address this issue, introducing bioactive drugs or factors is the most commonly used method, but there are often many problems such as high cost, uncontrollable and monotonous drug activity, and poor bioavailability. Here, a drug-free 3D printing PLLA scaffold with a triple-effect combination induced by surface-modified copper-doped layered double hydroxides (Cu-LDHs) is proposed. In the early stage of scaffold implantation, Cu-LDHs exert a photothermal therapy (PTT) effect to generate high temperature to effectively prevent bacterial infection. In the later stage, Cu-LDHs can further have a mild hyperthermia (MHT) effect to stimulate angiogenesis and osteogenic differentiation, demonstrating excellent vascularization and osteogenic activity. More importantly, with the degradation of Cu-LDHs, the released Cu2+ and Mg2+ provide an ion microenvironment effect and further synergize with the MHT effect to stimulate angiogenesis and osteogenic differentiation, thus more effectively promoting the healing of bone tissue. This triple-effect combined scaffold exhibits outstanding antibacterial, osteogenic, and angiogenic activities, as well as the advantages of low cost, convenient procedure, and long-term efficacy, and is expected to provide a promising strategy for clinical repair of bone defects.
Collapse
Affiliation(s)
- Zelin Zhu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Yating Lin
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Lin Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Kun Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Wei Wen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| | - Shan Ding
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| | - Mingxian Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| | - Lu Lu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| | - Changren Zhou
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| | - Binghong Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| |
Collapse
|
29
|
Han X, Saiding Q, Cai X, Xiao Y, Wang P, Cai Z, Gong X, Gong W, Zhang X, Cui W. Intelligent Vascularized 3D/4D/5D/6D-Printed Tissue Scaffolds. NANO-MICRO LETTERS 2023; 15:239. [PMID: 37907770 PMCID: PMC10618155 DOI: 10.1007/s40820-023-01187-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023]
Abstract
Blood vessels are essential for nutrient and oxygen delivery and waste removal. Scaffold-repairing materials with functional vascular networks are widely used in bone tissue engineering. Additive manufacturing is a manufacturing technology that creates three-dimensional solids by stacking substances layer by layer, mainly including but not limited to 3D printing, but also 4D printing, 5D printing and 6D printing. It can be effectively combined with vascularization to meet the needs of vascularized tissue scaffolds by precisely tuning the mechanical structure and biological properties of smart vascular scaffolds. Herein, the development of neovascularization to vascularization to bone tissue engineering is systematically discussed in terms of the importance of vascularization to the tissue. Additionally, the research progress and future prospects of vascularized 3D printed scaffold materials are highlighted and presented in four categories: functional vascularized 3D printed scaffolds, cell-based vascularized 3D printed scaffolds, vascularized 3D printed scaffolds loaded with specific carriers and bionic vascularized 3D printed scaffolds. Finally, a brief review of vascularized additive manufacturing-tissue scaffolds in related tissues such as the vascular tissue engineering, cardiovascular system, skeletal muscle, soft tissue and a discussion of the challenges and development efforts leading to significant advances in intelligent vascularized tissue regeneration is presented.
Collapse
Affiliation(s)
- Xiaoyu Han
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Qimanguli Saiding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
| | - Xiaolu Cai
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, People's Republic of China
| | - Yi Xiao
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Peng Wang
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Zhengwei Cai
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
| | - Xuan Gong
- University of Texas Southwestern Medical Center, Dallas, TX, 75390-9096, USA
| | - Weiming Gong
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China.
| | - Xingcai Zhang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
30
|
Gu Y, Zou Y, Huang Y, Liang R, Wu Y, Hu Y, Hong Y, Zhang X, Toh YC, Ouyang H, Zhang S. 3D-printed biomimetic scaffolds with precisely controlled and tunable structures guide cell migration and promote regeneration of osteochondral defect. Biofabrication 2023; 16:015003. [PMID: 37797606 DOI: 10.1088/1758-5090/ad0071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/05/2023] [Indexed: 10/07/2023]
Abstract
Untreated osteochondral defects will develop into osteoarthritis, affecting patients' quality of life. Since articular cartilage and subchondral bone exhibit distinct biological characteristics, repairing osteochondral defects remains a major challenge. Previous studies have tried to fabricate multilayer scaffolds with traditional methods or 3D printing technology. However, the efficacy is unsatisfactory because of poor control over internal structures or a lack of integrity between adjacent layers, severely compromising repair outcomes. Therefore, there is a need for a biomimetic scaffold that can simultaneously boost osteochondral defect regeneration in both structure and function. Herein, an integrated bilayer scaffold with precisely controlled structures is successfully 3D-printed in one step via digital light processing (DLP) technology. The upper layer has both 'lotus- and radial-' distribution pores, and the bottom layer has 'lotus-' pores to guide and facilitate the migration of chondrocytes and bone marrow mesenchymal stem cells, respectively, to the defect area. Tuning pore sizes could modulate the mechanical properties of scaffolds easily. Results show that 3D-printed porous structures allow significantly more cells to infiltrate into the area of 'lotus- and radial-' distribution pores during cell migration assay, subcutaneous implantation, andin situtransplantation, which are essential for osteochondral repair. Transplantation of this 3D-printed bilayer scaffold exhibits a promising osteochondral repair effect in rabbits. Incorporation of Kartogenin into the upper layer of scaffolds further induces better cartilage formation. Combining small molecules/drugs and precisely size-controlled and layer-specific porous structure via DLP technology, this 3D-printed bilayer scaffold is expected to be a potential strategy for osteochondral regeneration.
Collapse
Affiliation(s)
- Yuqing Gu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yiwei Zou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yuxuan Huang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Renjie Liang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yicong Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yifan Hu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yi Hong
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Xianzhu Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane 4059, Australia
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, People's Republic of China
- Department of Sports Medicine, School of Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, People's Republic of China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, People's Republic of China
| |
Collapse
|
31
|
Schoonraad SA, Jaimes AA, Singh AJX, Croland KJ, Bryant SJ. Osteogenic effects of covalently tethered rhBMP-2 and rhBMP-9 in an MMP-sensitive PEG hydrogel nanocomposite. Acta Biomater 2023; 170:53-67. [PMID: 37634836 PMCID: PMC10831697 DOI: 10.1016/j.actbio.2023.08.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
While bone morphogenic protein-2 (BMP-2) is one of the most widely studied BMPs in bone tissue engineering, BMP-9 has been purported to be a highly osteogenic BMP. This work investigates the individual osteogenic effects of recombinant human (rh) BMP-2 and rhBMP-9, when tethered into a hydrogel, on encapsulated human mesenchymal stem cells (MSCs). A matrix-metalloproteinase (MMP)-sensitive hydrogel nanocomposite, comprised of poly(ethylene glycol) crosslinked with MMP-sensitive peptides, tethered RGD, and entrapped hydroxyapatite nanoparticles was used. The rhBMPs were functionalized with free thiols and then covalently tethered into the hydrogel by a thiol-norbornene photoclick reaction. rhBMP-2 retained its full bioactivity post-thiolation, while the bioactivity of rhBMP-9 was partially reduced. Nonetheless, both rhBMPs were highly effective at enhancing osteogenesis over 12-weeks in a chemically-defined medium. Expression of ID1 and osterix, early markers of osteogenesis; collagen type I, a main component of the bone extracellular matrix (ECM); and osteopontin, bone sialoprotein II and dentin matrix protein I, mature osteoblast markers, increased with increasing concentrations of tethered rhBMP-2 or rhBMP-9. When comparing the two BMPs, rhBMP-9 led to more rapid collagen deposition and greater mineralization long-term. In summary, rhBMP-2 retained its bioactivity post-thiolation while rhBMP-9 is more susceptible to thiolation. Despite this shortcoming with rhBMP-9, both rhBMPs when tethered into this hydrogel, enhanced osteogenesis of MSCs, leading to a mature osteoblast phenotype surrounded by a mineralized ECM. STATEMENT OF SIGNIFICANCE: Osteoinductive hydrogels are a promising vehicle to deliver mesenchymal stem cells (MSCs) for bone regeneration. This study examines the in vitro osteoinductive capabilities when tethered bone morphogenic proteins (BMPs) are incorporated into a degradable biomimetic hydrogel with cell adhesive ligands, matrix metalloproteinase sensitive crosslinks for cell-mediated degradation, and hydroxyapatite nanoparticles. This study demonstrates that BMP-2 is readily thiolated and tethered without loss of bioactivity while bioactivity of BMP-9 is more susceptible to immobilization. Nonetheless, when either BMP2 or BMP9 are tethered into this hydrogel, osteogenesis of human MSCs is enhanced, bone extracellular matrix is deposited, and a mature osteoblast phenotype is achieved. This bone-biomimetic hydrogel is a promising design for stem cell-mediated bone regeneration.
Collapse
Affiliation(s)
- Sarah A Schoonraad
- Materials Science & Engineering Program, University of Colorado, 4001 Discovery Dr, Boulder, CO 80309-0613, United States
| | - Alan A Jaimes
- Department of Biochemistry, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, United States
| | - Arjun J X Singh
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, United States
| | - Kiera J Croland
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, United States
| | - Stephanie J Bryant
- Materials Science & Engineering Program, University of Colorado, 4001 Discovery Dr, Boulder, CO 80309-0613, United States; Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, United States; BioFrontiers Institute, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, United States.
| |
Collapse
|
32
|
Jia W, Zhou Z, Zhan W. Musculoskeletal Biomaterials: Stimulated and Synergized with Low Intensity Pulsed Ultrasound. J Funct Biomater 2023; 14:504. [PMID: 37888169 PMCID: PMC10607075 DOI: 10.3390/jfb14100504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Clinical biophysical stimulating strategies, which have significant effects on improving the function of organs or treating diseases by causing the salutary response of body, have shown many advantages, such as non-invasiveness, few side effects, and controllable treatment process. As a critical technique for stimulation, the low intensity pulsed ultrasound (LIPUS) has been explored in regulating osteogenesis, which has presented great promise in bone repair by delivering a combined effect with biomaterials. This review summarizes the musculoskeletal biomaterials that can be synergized with LIPUS for enhanced biomedical application, including bone regeneration, spinal fusion, osteonecrosis/osteolysis, cartilage repair, and nerve regeneration. Different types of biomaterials are categorized for summary and evaluation. In each subtype, the verified biological mechanisms are listed in a table or graphs to prove how LIPUS was effective in improving musculoskeletal tissue regeneration. Meanwhile, the acoustic excitation parameters of LIPUS that were promising to be effective for further musculoskeletal tissue engineering are discussed, as well as their limitations and some perspectives for future research. Overall, coupled with biomimetic scaffolds and platforms, LIPUS may be a powerful therapeutic approach to accelerate musculoskeletal tissue repair and even in other regenerative medicine applications.
Collapse
Affiliation(s)
- Wanru Jia
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Zifei Zhou
- Department of Orthopedics, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiwei Zhan
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| |
Collapse
|
33
|
Tan Y, Fan S, Wu X, Liu M, Dai T, Liu C, Ni S, Wang J, Yuan X, Zhao H, Weng Y. Fabrication of a three-dimensional printed gelatin/sodium alginate/nano-attapulgite composite polymer scaffold loaded with leonurine hydrochloride and its effects on osteogenesis and vascularization. Int J Biol Macromol 2023; 249:126028. [PMID: 37506787 DOI: 10.1016/j.ijbiomac.2023.126028] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
Bone tissue engineering scaffolds have made significant progress in treating bone defects in recent decades. However, the lack of a vascular network within the scaffold limits bone formation after implantation in vivo. Recent research suggests that leonurine hydrochloride (LH) can promote healing in full-thickness cutaneous wounds by increasing vessel formation and collagen deposition. Gelatin and Sodium Alginate are both polymers. ATP is a magnesium silicate chain mineral. In this study, a Gelatin/Sodium Alginate/Nano-Attapulgite composite hydrogel was used as the base material first, and the Gelatin/Sodium Alginate/Nano-Attapulgite composite polymer scaffold loaded with LH was then created using 3D printing technology. Finally, LH was grafted onto the base material by an amide reaction to construct a scaffold loaded with LH to achieve long-term LH release. When compared to pure polymer scaffolds, in vitro results showed that LH-loaded scaffolds promoted the differentiation of BMSCs into osteoblasts, as evidenced by increased expression of osteogenic key genes. The results of in vivo tissue staining revealed that the drug-loaded scaffold promoted both angiogenesis and bone formation. Collectively, these findings suggest that LH-loaded Gelatin/Sodium Alginate/Nano-Attapulgite composite hydrogel scaffolds are a potential therapeutic strategy and can assist bone regeneration.
Collapse
Affiliation(s)
- Yadong Tan
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Shijie Fan
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Xiaoyu Wu
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Menggege Liu
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Ting Dai
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Chun Liu
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Su Ni
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Jiafeng Wang
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Xiuchen Yuan
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Hongbin Zhao
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China.
| | - Yiping Weng
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213164, China; Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China.
| |
Collapse
|
34
|
Chen J, Zhou H, Fan Y, Gao G, Ying Y, Li J. 3D printing for bone repair: Coupling infection therapy and defect regeneration. CHEMICAL ENGINEERING JOURNAL 2023; 471:144537. [DOI: 10.1016/j.cej.2023.144537] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
|
35
|
Nagendla NK, Muralidharan K, Raju M, Mohan H, Selvakumar P, Bhandi MM, Mudiam MKR, Ramalingam V. Comprehensive metabolomic analysis of Mangifera indica leaves using UPLC-ESI-Q-TOF-MS E for cell differentiation: An in vitro and in vivo study. Food Res Int 2023; 171:112993. [PMID: 37330843 DOI: 10.1016/j.foodres.2023.112993] [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: 12/08/2022] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 06/19/2023]
Abstract
The comprehensive metabolic profiling was performed in the leaf extracts of Mangifera indica and assessed for their significant therapeutic application in tissue engineering and regenerative medicine in both in vitro and in vivo studies. About 147 compounds were identified in the ethyl acetate and methanol extracts of M. indica using MS/MS fragmentation analysis and the selected compounds were quantified using LC-QqQ-MS analysis. The in vitro cytotoxic activity showed that the M. indica extracts enhance the proliferation of mouse myoblast cells in concentration-dependent manner. As well, the extracts of M. indica induce the myotube formation by generating oxidative stress in the C2C12 cells was confirmed. The western blot analysis clearly showed that the M. indica induce myogenic differentiation by upregulating the myogenic marker proteins such as PI3K, Akt, mTOR, MyoG, and MyoD. The in vivo studies showed that the extracts expedites the acute wound repair by formation of crust, wound closure and improves the blood perfusion towards the wound area. Together, the leaves of M. indica can be used as excellent therapeutic agent for tissue repair and wound healing applications.
Collapse
Affiliation(s)
- Narendra Kumar Nagendla
- Department of Analytical & Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Kathirvel Muralidharan
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India; Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Malothu Raju
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India; Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Harshavardhan Mohan
- Department of Chemistry, Research Institute of Physics and Chemistry, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Piramanayagam Selvakumar
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Murali Mohan Bhandi
- Department of Analytical & Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Mohana Krishna Reddy Mudiam
- Department of Analytical & Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India.
| | - Vaikundamoorthy Ramalingam
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India; Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
| |
Collapse
|
36
|
Li X, Sung P, Zhang D, Yan L. Curcumin in vitro Neuroprotective Effects Are Mediated by p62/keap-1/Nrf2 and PI3K/AKT Signaling Pathway and Autophagy Inhibition. Physiol Res 2023; 72:497-510. [PMID: 37795892 PMCID: PMC10634561 DOI: 10.33549/physiolres.935054] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/11/2023] [Indexed: 01/05/2024] Open
Abstract
Oxidative stress and autophagy are potential mechanisms associated with cerebral ischemia/reperfusion injury (IRI) and is usually linked to inflammatory responses and apoptosis. Curcumin has recently been demonstrated to exhibit anti-inflammatory, anti-oxidant, anti-apoptotic and autophagy regulation properties. However, mechanism of curcumin on IRI-induced oxidative stress and autophagy remains not well understood. We evaluated the protective effects and potential mechanisms of curcumin on cerebral microvascular endothelial cells (bEnd.3) and neuronal cells (HT22) against oxygen glucose deprivation/reoxygenation (OGD/R) in vitro models that mimic in vivo cerebral IRI. The cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) activity assays revealed that curcumin attenuated the OGD/R-induced injury in a dose-specific manner. OGD/R induced elevated levels of inflammatory cytokines TNF-alpha, IL-6 as well as IL-1beta, and these effects were notably reduced by curcumin. OGD/R-mediated apoptosis was suppressed by curcumin via upregulating B-cell lymphoma-2 (Bcl-2) and downregulating Bcl-associated X (Bax), cleaved-caspase3 and TUNEL apoptosis marker. Additionally, curcumin increased superoxide dismutase (SOD) and glutathione (GSH), but suppressed malondialdehyde (MDA) and reactive oxygen species (ROS) content. Curcumin inhibited the levels of autophagic biomarkers such as LC3 II/LC3 I and Beclin1. Particularly, curcumin induced p62 accumulation and its interactions with keap1 and promoted NF-E2-related factor 2 (Nrf2) translocation to nucleus, accompanied by increased NADPH quinone dehydrogenase (Nqo1) and heme oxygenase 1 (HO-1). Treatment of curcumin increased phosphorylation-phosphatidylinositol 3 kinase (p-PI3K) and p-protein kinase B (p-AKT). The autophagy inhibitor 3-methyladenine (3-MA) activated the keap-1/Nrf2 and PI3K/AKT pathways. This study highlights the neuroprotective effects of curcumin on cerebral IRI.
Collapse
Affiliation(s)
- X Li
- Department of Neurology, Tangshan Gongren Hospital, Tangshan, Hebei Province, China
| | | | | | | |
Collapse
|
37
|
Liu X, Gao J, Cui X, Nie S, Wu X, Zhang L, Tang P, Liu J, Li M. Functionalized 3D-Printed PLA Biomimetic Scaffold for Repairing Critical-Size Bone Defects. Bioengineering (Basel) 2023; 10:1019. [PMID: 37760121 PMCID: PMC10526104 DOI: 10.3390/bioengineering10091019] [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: 07/04/2023] [Revised: 08/04/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
The treatment of critical-size bone defects remains a complicated clinical challenge. Recently, bone tissue engineering has emerged as a potential therapeutic approach for defect repair. This study examined the biocompatibility and repair efficacy of hydroxyapatite-mineralized bionic polylactic acid (PLA) scaffolds, which were prepared through a combination of 3D printing technology, plasma modification, collagen coating, and hydroxyapatite mineralization coating techniques. Physicochemical analysis, mechanical testing, and in vitro and animal experiments were conducted to elucidate the impact of structural design and microenvironment on osteogenesis. Results indicated that the PLA scaffold exhibited a porosity of 84.1% and a pore size of 350 μm, and its macrostructure was maintained following functionalization modification. The functionalized scaffold demonstrated favorable hydrophilicity and biocompatibility and promoted cell adhesion, proliferation, and the expression of osteogenic genes such as ALP, OPN, Col-1, OCN, and RUNX2. Moreover, the scaffold was able to effectively repair critical-size bone defects in the rabbit radius, suggesting a novel strategy for the treatment of critical-size bone defects.
Collapse
Affiliation(s)
- Xiao Liu
- Medical School of Chinese PLA, Beijing 100853, China; (X.L.); (J.G.)
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; (X.C.); (S.N.); (X.W.); (L.Z.); (P.T.)
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Jianpeng Gao
- Medical School of Chinese PLA, Beijing 100853, China; (X.L.); (J.G.)
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; (X.C.); (S.N.); (X.W.); (L.Z.); (P.T.)
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Xiang Cui
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; (X.C.); (S.N.); (X.W.); (L.Z.); (P.T.)
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Shaobo Nie
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; (X.C.); (S.N.); (X.W.); (L.Z.); (P.T.)
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Xiaoyong Wu
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; (X.C.); (S.N.); (X.W.); (L.Z.); (P.T.)
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Licheng Zhang
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; (X.C.); (S.N.); (X.W.); (L.Z.); (P.T.)
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Peifu Tang
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; (X.C.); (S.N.); (X.W.); (L.Z.); (P.T.)
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Jianheng Liu
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; (X.C.); (S.N.); (X.W.); (L.Z.); (P.T.)
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Ming Li
- Department of Orthopaedics, The Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; (X.C.); (S.N.); (X.W.); (L.Z.); (P.T.)
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| |
Collapse
|
38
|
Guo X, Song P, Li F, Yan Q, Bai Y, He J, Che Q, Cao H, Guo J, Su Z. Research Progress of Design Drugs and Composite Biomaterials in Bone Tissue Engineering. Int J Nanomedicine 2023; 18:3595-3622. [PMID: 37416848 PMCID: PMC10321437 DOI: 10.2147/ijn.s415666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/13/2023] [Indexed: 07/08/2023] Open
Abstract
Bone, like most organs, has the ability to heal naturally and can be repaired slowly when it is slightly injured. However, in the case of bone defects caused by diseases or large shocks, surgical intervention and treatment of bone substitutes are needed, and drugs are actively matched to promote osteogenesis or prevent infection. Oral administration or injection for systemic therapy is a common way of administration in clinic, although it is not suitable for the long treatment cycle of bone tissue, and the drugs cannot exert the greatest effect or even produce toxic and side effects. In order to solve this problem, the structure or carrier simulating natural bone tissue is constructed to control the loading or release of the preparation with osteogenic potential, thus accelerating the repair of bone defect. Bioactive materials provide potential advantages for bone tissue regeneration, such as physical support, cell coverage and growth factors. In this review, we discuss the application of bone scaffolds with different structural characteristics made of polymers, ceramics and other composite materials in bone regeneration engineering and drug release, and look forward to its prospect.
Collapse
Affiliation(s)
- Xinghua Guo
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
| | - Pan Song
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
| | - Feng Li
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
| | - Qihao Yan
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510310, People’s Republic of China
| | - Jincan He
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510310, People’s Republic of China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd, Science City, Guangzhou, 510663, People’s Republic of China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, 528458, People’s Republic of China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
| |
Collapse
|
39
|
Laubach M, Hildebrand F, Suresh S, Wagels M, Kobbe P, Gilbert F, Kneser U, Holzapfel BM, Hutmacher DW. The Concept of Scaffold-Guided Bone Regeneration for the Treatment of Long Bone Defects: Current Clinical Application and Future Perspective. J Funct Biomater 2023; 14:341. [PMID: 37504836 PMCID: PMC10381286 DOI: 10.3390/jfb14070341] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/31/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
The treatment of bone defects remains a challenging clinical problem with high reintervention rates, morbidity, and resulting significant healthcare costs. Surgical techniques are constantly evolving, but outcomes can be influenced by several parameters, including the patient's age, comorbidities, systemic disorders, the anatomical location of the defect, and the surgeon's preference and experience. The most used therapeutic modalities for the regeneration of long bone defects include distraction osteogenesis (bone transport), free vascularized fibular grafts, the Masquelet technique, allograft, and (arthroplasty with) mega-prostheses. Over the past 25 years, three-dimensional (3D) printing, a breakthrough layer-by-layer manufacturing technology that produces final parts directly from 3D model data, has taken off and transformed the treatment of bone defects by enabling personalized therapies with highly porous 3D-printed implants tailored to the patient. Therefore, to reduce the morbidities and complications associated with current treatment regimens, efforts have been made in translational research toward 3D-printed scaffolds to facilitate bone regeneration. Three-dimensional printed scaffolds should not only provide osteoconductive surfaces for cell attachment and subsequent bone formation but also provide physical support and containment of bone graft material during the regeneration process, enhancing bone ingrowth, while simultaneously, orthopaedic implants supply mechanical strength with rigid, stable external and/or internal fixation. In this perspective review, we focus on elaborating on the history of bone defect treatment methods and assessing current treatment approaches as well as recent developments, including existing evidence on the advantages and disadvantages of 3D-printed scaffolds for bone defect regeneration. Furthermore, it is evident that the regulatory framework and organization and financing of evidence-based clinical trials remains very complex, and new challenges for non-biodegradable and biodegradable 3D-printed scaffolds for bone regeneration are emerging that have not yet been sufficiently addressed, such as guideline development for specific surgical indications, clinically feasible design concepts for needed multicentre international preclinical and clinical trials, the current medico-legal status, and reimbursement. These challenges underscore the need for intensive exchange and open and honest debate among leaders in the field. This goal can be addressed in a well-planned and focused stakeholder workshop on the topic of patient-specific 3D-printed scaffolds for long bone defect regeneration, as proposed in this perspective review.
Collapse
Affiliation(s)
- Markus Laubach
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Frank Hildebrand
- Department of Orthopaedics, Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Sinduja Suresh
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Michael Wagels
- Department of Plastic Surgery, Princess Alexandra Hospital, Woolloongabba, QLD 4102, Australia;
- The Herston Biofabrication Institute, The University of Queensland, Herston, QLD 4006, Australia
- Southside Clinical Division, School of Medicine, University of Queensland, Woolloongabba, QLD 4102, Australia
- Department of Plastic and Reconstructive Surgery, Queensland Children’s Hospital, South Brisbane, QLD 4101, Australia
- The Australian Centre for Complex Integrated Surgical Solutions, Woolloongabba, QLD 4102, Australia
| | - Philipp Kobbe
- Department of Orthopaedics, Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Fabian Gilbert
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Ulrich Kneser
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, 67071 Ludwigshafen, Germany
| | - Boris M. Holzapfel
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Dietmar W. Hutmacher
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, QLD 4000, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies (CTET), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| |
Collapse
|
40
|
Jo Y, Sarkar N, Bose S. In vitro biological evaluation of epigallocatechin gallate (EGCG) release from three-dimensional printed (3DP) calcium phosphate bone scaffolds. J Mater Chem B 2023; 11:5503-5513. [PMID: 36637404 PMCID: PMC11132590 DOI: 10.1039/d2tb02210a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Three-dimensional printed (3DP) tricalcium phosphate (TCP) scaffolds can guide bone regeneration, especially for patient-specific defect repair applications in low-load bearing sites. Epigallocatechin gallate (EGCG), a green tea compound, has gained attention as a safer alternative treatment for bone disorders. The 3DP TCP scaffold is designed for localized EGCG delivery, which can enhance in vitro osteogenic ability, anti-osteoclastogenic activity, vascularization formation, and chemoprevention. In the cocultures of human bone marrow-derived mesenchymal stem cells (hMSCs) and monocytes (THP-1), EGCG release enhances osteogenic differentiation of hMSCs at day 16 compared to the control; this is indicated by a 2.8- and 4.0-fold upregulation of Runt-related transcription factor 2 (Runx2) and bone gamma-carboxyglutamic acid-containing protein (BGLAP), the early and late osteoblast differentiation marker expressions. However, EGCG significantly downregulates the receptor activator of nuclear factor-κB ligand (RANKL) expression by 7.0-fold, indicating that EGCG suppresses RANKL-induced osteoclast maturation. EGCG also stimulates endothelial tube formation at as early as 3 hours when human umbilical vein endothelial cells (HUVECs) grow on Matrigel. It reduces human osteosarcoma MG-63 cell viability by 66% compared to the control at day 11. An in vitro release kinetics study demonstrates that EGCG shows a ∼64% release within a day followed by a sustained release in the physiological environment (pH 7.4) because its phenolic hydroxyl groups are easily deprotonated at physiological pH. These findings contribute to developing a multifunctional scaffold for the treatment of low load-bearing patient-specific bone defects after trauma and tumor excision.
Collapse
Affiliation(s)
- Yongdeok Jo
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, USA.
| | - Naboneeta Sarkar
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, USA.
| | - Susmita Bose
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, USA.
| |
Collapse
|
41
|
Yang Z, Li H, Lin J, Xing D, Li JJ, Cribbin EM, Kim AM, He Z, Li H, Guo W, Zhang L, Lin J. Research landscape of 3D printing in bone regeneration and bone repair: A bibliometric and visualized analysis from 2012 to 2022. Int J Bioprint 2023; 9:737. [PMID: 37323492 PMCID: PMC10261130 DOI: 10.18063/ijb.737] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/24/2023] [Indexed: 06/17/2023] Open
Abstract
Three-dimensional printing (3DP) is a popular manufacturing technique with versatile potential for materials processing in tissue engineering and regenerative medicine. In particular, the repair and regeneration of significant bone defects remain as substantial clinical challenges that require biomaterial implants to maintain mechanical strength and porosity, which may be realized using 3DP. The rapid progress in 3DP development in the past decade warrants a bibliometric analysis to gain insights into its applications in bone tissue engineering (BTE). Here, we performed a comparative study using bibliometric methods for 3DP in bone repair and regeneration. A total of 2,025 articles were included, and the results showed an increase in the number of publications and relative research interest on 3DP annually worldwide. China was the leader in international cooperation in this field and also the largest contributor to the number of citations. The majority of articles in this field were published in the journal Biofabrication. Chen Y was the author who made the highest contribution to the included studies. The keywords included in the publications were mainly related to BTE and regenerative medicine (including "3DP techniques," "3DP materials," "bone regeneration strategies," and "bone disease therapeutics") for bone regeneration and repair. This bibliometric and visualized analysis provides significant insights into the historical development of 3DP in BTE from 2012 to 2022, which will be beneficial for scientists to conduct further investigations into this dynamic field.
Collapse
Affiliation(s)
- Zhen Yang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Hao Li
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Dan Xing
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, Australia
| | - Elise M. Cribbin
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, Australia
| | - Alice M. Kim
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, Australia
| | - Zihao He
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Hui Li
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Weimin Guo
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jianhao Lin
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| |
Collapse
|
42
|
Predoi D, Iconaru SL, Ciobanu CS, Raita MS, Ghegoiu L, Trusca R, Badea ML, Cimpeanu C. Studies of the Tarragon Essential Oil Effects on the Characteristics of Doped Hydroxyapatite/Chitosan Biocomposites. Polymers (Basel) 2023; 15:polym15081908. [PMID: 37112055 PMCID: PMC10142985 DOI: 10.3390/polym15081908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Due to the emergence of antibiotic-resistant pathogens, the need to find new, efficient antimicrobial agents is rapidly increasing. Therefore, in this study, we report the development of new biocomposites based on zinc-doped hydroxyapatite/chitosan enriched with essential oil of Artemisia dracunculus L. with good antimicrobial activity. Techniques such as scanning electron microscopy (SEM), X-ray diffraction (XRD), energy dispersive X-ray spectroscopy (EDX) and Fourier transform infrared spectroscopy (FTIR) were used in order to evaluate their physico-chemical properties. Our studies revealed that biocomposite materials with nanometric dimension and homogeneous composition could be obtained through an economic and cost-effective synthesis method. The biological assays demonstrated that ZnHA (zinc-doped hydroxyapatite), ZnHACh (zinc-doped hydroxyapatite/chitosan) and ZnHAChT (zinc-doped hydroxyapatite/chitosan enriched with essential oil of Artemisia dracunculus L.) did not exhibit a toxic effect on the cell viability and proliferation of the primary osteoblast culture (hFOB 1.19). Moreover, the cytotoxic assay also highlighted that the cell morphology of the hFOB 1.19 was not altered in the presence of ZnHA, ZnHACh or ZnHAChT. Furthermore, the in vitro antimicrobial studies emphasized that the samples exhibited strong antimicrobial properties against Escherichia coli ATCC 25922, Staphylococcus aureus ATCC 25923 and Candida albicans ATCC 10231 microbial strains. These results are encouraging for the following development of new composite materials with enhanced biological properties that could promote the osteogenic process of bone healing and also exhibit good antimicrobial properties.
Collapse
Affiliation(s)
- Daniela Predoi
- National Institute of Materials Physics, Atomistilor Street, No. 405A, 077125 Magurele, Romania
| | - Simona Liliana Iconaru
- National Institute of Materials Physics, Atomistilor Street, No. 405A, 077125 Magurele, Romania
| | - Carmen Steluta Ciobanu
- National Institute of Materials Physics, Atomistilor Street, No. 405A, 077125 Magurele, Romania
| | - Mariana Stefania Raita
- Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 105 Splaiul Independentei, District 5, 050097 Bucharest, Romania
| | - Liliana Ghegoiu
- National Institute of Materials Physics, Atomistilor Street, No. 405A, 077125 Magurele, Romania
| | - Roxana Trusca
- Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University POLITEHNICA of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
- National Center for Micro and Nanomaterials, University POLITEHNICA of Bucharest, Splaiul Independentei 313, 060042 Bucharest, Romania
| | - Monica Luminita Badea
- Faculty of Horticulture, University of Agronomic Sciences and Veterinary Medicine, 59 Marasti Blvd., 011464 Bucharest, Romania
| | - Carmen Cimpeanu
- Faculty of Land Reclamation and Environmental Engineering, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 59 Marasti Blvd, 011464 Bucharest, Romania
| |
Collapse
|
43
|
Recent advances in carboxymethyl chitosan-based materials for biomedical applications. Carbohydr Polym 2023; 305:120555. [PMID: 36737218 DOI: 10.1016/j.carbpol.2023.120555] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/12/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Chitosan (CS) and its derivatives have been applied extensively in the biomedical field owing to advantageous characteristics including biodegradability, biocompatibility, antibacterial activity and adhesive properties. The low solubility of CS at physiological pH limits its use in systems requiring higher dissolving ability and a suitable drug release rate. Besides, CS can result in fast drug release because of its high swelling degree and rapid water absorption in aqueous media. As a water-soluble derivative of CS, carboxymethyl chitosan (CMC) has certain improved properties, rendering it a more suitable candidate for wound healing, drug delivery and tissue engineering applications. This review will focus on the antibacterial, anticancer and antitumor, antioxidant and antifungal bioactivities of CMC and the most recently described applications of CMC in wound healing, drug delivery, tissue engineering, bioimaging and cosmetics.
Collapse
|
44
|
Application of 3D Printing in Bone Grafts. Cells 2023; 12:cells12060859. [PMID: 36980200 PMCID: PMC10047278 DOI: 10.3390/cells12060859] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
The application of 3D printing in bone grafts is gaining in importance and is becoming more and more popular. The choice of the method has a direct impact on the preparation of the patient for surgery, the probability of rejection of the transplant, and many other complications. The aim of the article is to discuss methods of bone grafting and to compare these methods. This review of literature is based on a selective literature search of the PubMed and Web of Science databases from 2001 to 2022 using the search terms “bone graft”, “bone transplant”, and “3D printing”. In addition, we also reviewed non-medical literature related to materials used for 3D printing. There are several methods of bone grafting, such as a demineralized bone matrix, cancellous allograft, nonvascular cortical allograft, osteoarticular allograft, osteochondral allograft, vascularized allograft, and an autogenic transplant using a bone substitute. Currently, autogenous grafting, which involves removing the patient’s bone from an area of low aesthetic importance, is referred to as the gold standard. 3D printing enables using a variety of materials. 3D technology is being applied to bone tissue engineering much more often. It allows for the treatment of bone defects thanks to the creation of a porous scaffold with adequate mechanical strength and favorable macro- and microstructures. Bone tissue engineering is an innovative approach that can be used to repair multiple bone defects in the process of transplantation. In this process, biomaterials are a very important factor in supporting regenerative cells and the regeneration of tissue. We have years of research ahead of us; however, it is certain that 3D printing is the future of transplant medicine.
Collapse
|
45
|
He Y, Li F, Jiang P, Cai F, Lin Q, Zhou M, Liu H, Yan F. Remote control of the recruitment and capture of endogenous stem cells by ultrasound for in situ repair of bone defects. Bioact Mater 2023; 21:223-238. [PMID: 36157244 PMCID: PMC9465026 DOI: 10.1016/j.bioactmat.2022.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Stem cell-based tissue engineering has provided a promising platform for repairing of bone defects. However, the use of exogenous bone marrow mesenchymal stem cells (BMSCs) still faces many challenges such as limited sources and potential risks. It is important to develop new approach to effectively recruit endogenous BMSCs and capture them for in situ bone regeneration. Here, we designed an acoustically responsive scaffold (ARS) and embedded it into SDF-1/BMP-2 loaded hydrogel to obtain biomimetic hydrogel scaffold complexes (BSC). The SDF-1/BMP-2 cytokines can be released on demand from the BSC implanted into the defected bone via pulsed ultrasound (p-US) irradiation at optimized acoustic parameters, recruiting the endogenous BMSCs to the bone defected or BSC site. Accompanied by the daily p-US irradiation for 14 days, the alginate hydrogel was degraded, resulting in the exposure of ARS to these recruited host stem cells. Then another set of sinusoidal continuous wave ultrasound (s-US) irradiation was applied to excite the ARS intrinsic resonance, forming highly localized acoustic field around its surface and generating enhanced acoustic trapping force, by which these recruited endogenous stem cells would be captured on the scaffold, greatly promoting them to adhesively grow for in situ bone tissue regeneration. Our study provides a novel and effective strategy for in situ bone defect repairing through acoustically manipulating endogenous BMSCs. We designed ARS and embedded it into SDF-1/BMP-2 loaded hydrogel to form BSC. The BSC can release SDF-1/BMP-2 by p-US irradiation for recruitment of endogenous BMSCs and capture them by s-US irradiation. The in situ repair of bone defects were successfully realized by US-mediated control of the recruitment and capture of BMSCs.
Collapse
Affiliation(s)
- Yanni He
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Peng Jiang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Feiyan Cai
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Qin Lin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Meijun Zhou
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
| | - Hongmei Liu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
- Corresponding author. Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China.
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
- Corresponding author. Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
46
|
Bandyopadhyay A, Mitra I, Goodman SB, Kumar M, Bose S. Improving Biocompatibility for Next Generation of Metallic Implants. PROGRESS IN MATERIALS SCIENCE 2023; 133:101053. [PMID: 36686623 PMCID: PMC9851385 DOI: 10.1016/j.pmatsci.2022.101053] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The increasing need for joint replacement surgeries, musculoskeletal repairs, and orthodontics worldwide prompts emerging technologies to evolve with healthcare's changing landscape. Metallic orthopaedic materials have a shared application history with the aerospace industry, making them only partly efficient in the biomedical domain. However, suitability of metallic materials in bone tissue replacements and regenerative therapies remains unchallenged due to their superior mechanical properties, eventhough they are not perfectly biocompatible. Therefore, exploring ways to improve biocompatibility is the most critical step toward designing the next generation of metallic biomaterials. This review discusses methods of improving biocompatibility of metals used in biomedical devices using surface modification, bulk modification, and incorporation of biologics. Our investigation spans multiple length scales, from bulk metals to the effect of microporosities, surface nanoarchitecture, and biomolecules such as DNA incorporation for enhanced biological response in metallic materials. We examine recent technologies such as 3D printing in alloy design and storing surface charge on nanoarchitecture surfaces, metal-on-metal, and ceramic-on-metal coatings to present a coherent and comprehensive understanding of the subject. Finally, we consider the advantages and challenges of metallic biomaterials and identify future directions.
Collapse
Affiliation(s)
- Amit Bandyopadhyay
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920
| | - Indranath Mitra
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920
| | - Stuart B. Goodman
- Department of Orthopedic Surgery, Stanford University Medical Center, Redwood City, CA 94063
| | | | - Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920
| |
Collapse
|
47
|
Bakhshandeh B, Ranjbar N, Abbasi A, Amiri E, Abedi A, Mehrabi M, Dehghani Z, Pennisi CP. Recent progress in the manipulation of biochemical and biophysical cues for engineering functional tissues. Bioeng Transl Med 2023; 8:e10383. [PMID: 36925674 PMCID: PMC10013802 DOI: 10.1002/btm2.10383] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/28/2022] [Accepted: 07/16/2022] [Indexed: 11/11/2022] Open
Abstract
Tissue engineering (TE) is currently considered a cutting-edge discipline that offers the potential for developing treatments for health conditions that negatively affect the quality of life. This interdisciplinary field typically involves the combination of cells, scaffolds, and appropriate induction factors for the regeneration and repair of damaged tissue. Cell fate decisions, such as survival, proliferation, or differentiation, critically depend on various biochemical and biophysical factors provided by the extracellular environment during developmental, physiological, and pathological processes. Therefore, understanding the mechanisms of action of these factors is critical to accurately mimic the complex architecture of the extracellular environment of living tissues and improve the efficiency of TE approaches. In this review, we recapitulate the effects that biochemical and biophysical induction factors have on various aspects of cell fate. While the role of biochemical factors, such as growth factors, small molecules, extracellular matrix (ECM) components, and cytokines, has been extensively studied in the context of TE applications, it is only recently that we have begun to understand the effects of biophysical signals such as surface topography, mechanical, and electrical signals. These biophysical cues could provide a more robust set of stimuli to manipulate cell signaling pathways during the formation of the engineered tissue. Furthermore, the simultaneous application of different types of signals appears to elicit synergistic responses that are likely to improve functional outcomes, which could help translate results into successful clinical therapies in the future.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Nika Ranjbar
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Elahe Amiri
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Ali Abedi
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Mohammad‐Reza Mehrabi
- Department of Microbial Biotechnology, School of Biology, College of ScienceUniversity of TehranTehranIran
| | - Zahra Dehghani
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and TechnologyAalborg UniversityAalborgDenmark
| |
Collapse
|
48
|
Gao J, Li M, Cheng J, Liu X, Liu Z, Liu J, Tang P. 3D-Printed GelMA/PEGDA/F127DA Scaffolds for Bone Regeneration. J Funct Biomater 2023; 14:jfb14020096. [PMID: 36826895 PMCID: PMC9962173 DOI: 10.3390/jfb14020096] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
Tissue-engineered scaffolds are an effective method for the treatment of bone defects, and their structure and function are essential for bone regeneration. Digital light processing (DLP) printing technology has been widely used in bone tissue engineering (BTE) due to its high printing resolution and gentle printing process. As commonly used bioinks, synthetic polymers such as polyethylene glycol diacrylate (PEGDA) and Pluronic F127 diacrylate (F127DA) have satisfactory printability and mechanical properties but usually lack sufficient adhesion to cells and tissues. Here, a compound BTE scaffold based on PEGDA, F127DA, and gelatin methacrylate (GelMA) was successfully prepared using DLP printing technology. The scaffold not only facilitated the adhesion and proliferation of cells, but also effectively promoted the osteogenic differentiation of mesenchymal stem cells in an osteoinductive environment. Moreover, the bone tissue volume/total tissue volume (BV/TV) of the GelMA/PEGDA/F127DA (GPF) scaffold in vivo was 49.75 ± 8.50%, higher than the value of 37.10 ± 7.27% for the PEGDA/F127DA (PF) scaffold and 20.43 ± 2.08% for the blank group. Therefore, the GPF scaffold prepared using DLP printing technology provides a new approach to the treatment of bone defects.
Collapse
Affiliation(s)
- Jianpeng Gao
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- Medical School of Chinese PLA, Beijing 100039, China
| | - Ming Li
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Junyao Cheng
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- Medical School of Chinese PLA, Beijing 100039, China
| | - Xiao Liu
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- Medical School of Chinese PLA, Beijing 100039, China
| | - Zhongyang Liu
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Jianheng Liu
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
- Correspondence: (J.L.); (P.T.)
| | - Peifu Tang
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
- Correspondence: (J.L.); (P.T.)
| |
Collapse
|
49
|
Ribeiro MEA, Checca Huaman NR, Gomez JGC, Rodríguez RJS. Poly(3-hydroxybutyrate-co-3-hydroxyvalerate) and amino-functionalized nanodiamond bionanocomposites for bone tissue defect repair. Int J Biol Macromol 2023; 226:1041-1053. [PMID: 36435460 DOI: 10.1016/j.ijbiomac.2022.11.221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Injection-molded nanocomposites of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBHV) with 6 % of 3-hydroxyvalerate (HV) and amino-nanodiamonds (nD-A) were produced and characterized to investigate the effect of functionalized nanodiamonds on mechanical and biological behavior to bone replacement application. To prepare mixtures of PHBHV and nD-A in different concentrations, nD-A was dispersed in chloroform by sonication with 40 % of amplitude. Three specimens were characterized by infrared spectroscopy (FTIR), thermogravimetric analysis (TGA), X-ray diffraction (DRX), differential scanning calorimetry (DSC), 3-point flexural tests, dynamic mechanical analysis (DMA), and scanning electron microscopy (SEM). FTIR and TGA evidenced the existence of interactions between the nD-A and PHBHV. The crystallinity degree of PHBHV slightly reduced (~9 %) in nanocomposites and the morphology of the crystals changed. Nanocomposites achieved satisfactory dispersion and distribution of nD-A for low concentrations. Elastic modulus (E) increased from 1.96 ± 0.20 (PHBHV) to 2.59 ± 0.19 GPa (PHBHV/1.0%nD-A) (30 %). Despite the relatively limited dispersion, PHBHV/2.0 % nD-A had the best combination of E, strength, and maximum deformation. It had the highest glass transition temperature (43.1 vs 40.3 °C of PHBHV) and the best adhesion coefficient and reinforcement effectiveness. PHBHV-nD-A did not induce toxicity in 7 days and allowed cell fixation and expansion. These bionanocomposites should be considered for supplementary studies for bone tissue engineering.
Collapse
Affiliation(s)
- Maria Eduarda Araújo Ribeiro
- Advanced Materials Laboratory-LAMAV, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Av. Alberto Lamego, 2000, Parque Califórnia, 28015-620 Campos dos Goytacazes, RJ, Brazil.
| | - Noemi Raquel Checca Huaman
- Centro Brasileiro de Pesquisas Físicas-CBPF, R. Dr. Xavier Sigaud, 150, 22290-180 Rio de Janeiro, RJ, Brazil
| | | | - Rubén J Sánchez Rodríguez
- Advanced Materials Laboratory-LAMAV, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Av. Alberto Lamego, 2000, Parque Califórnia, 28015-620 Campos dos Goytacazes, RJ, Brazil
| |
Collapse
|
50
|
Mirkhalaf M, Men Y, Wang R, No Y, Zreiqat H. Personalized 3D printed bone scaffolds: A review. Acta Biomater 2023; 156:110-124. [PMID: 35429670 DOI: 10.1016/j.actbio.2022.04.014] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/23/2022] [Accepted: 04/07/2022] [Indexed: 01/18/2023]
Abstract
3D printed bone scaffolds have the potential to replace autografts and allografts because of advantages such as unlimited supply and the ability to tailor the scaffolds' biochemical, biological and biophysical properties. Significant progress has been made over the past decade in additive manufacturing techniques to 3D print bone grafts, but challenges remain in the lack of manufacturing techniques that can recapitulate both mechanical and biological functions of native bones. The purpose of this review is to outline the recent progress and challenges of engineering an ideal synthetic bone scaffold and to provide suggestions for overcoming these challenges through bioinspiration, high-resolution 3D printing, and advanced modeling techniques. The article provides a short overview of the progress in developing the 3D printed scaffolds for the repair and regeneration of critical size bone defects. STATEMENT OF SIGNIFICANCE: Treatment of critical size bone defects is still a tremendous clinical challenge. To address this challenge, diverse sets of advanced manufacturing approaches and materials have been developed for bone tissue scaffolds. 3D printing has sparked much interest because it provides a close control over the scaffold's internal architecture and in turn its mechanical and biological properties. This article provides a critical overview of the relationships between material compositions, printing techniques, and properties of the scaffolds and discusses the current technical challenges facing their successful translation to the clinic. Bioinspiration, high-resolution printing, and advanced modeling techniques are discussed as future directions to address the current challenges.
Collapse
Affiliation(s)
- Mohammad Mirkhalaf
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia; Australian Research Council Training Centre for Innovative Bioengineering, Sydney, NSW 2006, Australia; School of Mechanical, Medical and Process Engineering, Queensland University of Technology, 2 George St., Brisbane, QLD 4000 Australia.
| | - Yinghui Men
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia
| | - Rui Wang
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia
| | - Young No
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia; Australian Research Council Training Centre for Innovative Bioengineering, Sydney, NSW 2006, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia; Australian Research Council Training Centre for Innovative Bioengineering, Sydney, NSW 2006, Australia.
| |
Collapse
|