1
|
Yu X, Zou Z, Li Y, Li J, Chen Y, Shi W, Liu X, Guo R, Cai X. Fiber-reinforced gelatin-based hydrogel biocomposite tubular scaffolds with programmable mechanical properties. Biomed Mater 2025; 20:035031. [PMID: 40306305 DOI: 10.1088/1748-605x/add2bc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/30/2025] [Indexed: 05/02/2025]
Abstract
Tissue-engineered tubular scaffolds (TETS) provide an effective repair solution for human tubular tissue loss and damage caused by congenital defects, disease, or mechanical trauma. However, there are still major challenges to developing TETS with excellent mechanical properties and biocompatibility for human tubular tissue repair. Gelatin-based hydrogels are suitable candidates for tissue-engineered scaffolds because they are hydrolyzed collagen products and have excellent biocompatibility and degradability. However, the mechanical properties of gelatin-based hydrogels are relatively poor and do not align well with the mechanical properties of human tubular tissues. Inspired by the extracellular matrix architecture of human tubular tissues, this study utilizes high-precision 3D printing to fabricate ultrafine fiber network tubular scaffolds (UFNTS) that mimic the arrangement of collagen fibers, which are then embedded in a cell-compatible gelatin-based hydrogel, resulting in the preparation of a fiber/hydrogel biocomposite tubular scaffold (BCTS) with tunable mechanical properties and a J-shaped stress-strain response. Finite element analysis was employed to predict the mechanical behavior of the UFNTS and BCTS. Experimental results indicate that by modifying the structural parameters of the UFNTS, the mechanical properties of the BCTS can be effectively tuned, achieving a programmable range of tensile modulus (0.2-4.35 MPa) and burst pressure (1580-7850 mmHg), which broadly covers the mechanical properties of most human tubular tissues. The design and fabrication of BCTS offer a new approach for the development of TETS while also providing a personalized strategy for such scaffolds in tissue engineering.
Collapse
Affiliation(s)
- Xiong Yu
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Zhongfei Zou
- School of Mechanical Engineering, Guizhou Institute of Technology, Guiyang 550003, People's Republic of China
| | - Yi Li
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Jiachun Li
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Yuewei Chen
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Wenhai Shi
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Xixia Liu
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Rui Guo
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Xianhui Cai
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| |
Collapse
|
2
|
Geng X, Yao Y, Huang H, Li Q, Wang L, Fan Y. Mechanical and biological characteristics of 3D-printed auxetic structure in bone tissue engineering. J Biomech 2025; 184:112685. [PMID: 40215656 DOI: 10.1016/j.jbiomech.2025.112685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
The auxetic structures are highly effective in bone implants due to their unique deformation characteristics. However, ideal tissue engineering scaffolds must possess suitable mechanical properties and biocompatibility. The biological effects of auxetic structures require further study. In this study, three types of 3D re-entrant honeycomb structures with varying angles of 75°, 90°, and 105° were designed. These structures were fabricated by stereolithography 3D printing technology. Finite element simulations and compression tests were conducted to evaluate their mechanical properties. Scaffolds were inoculated with preosteoblast MC3T3-E1 cells, and cyclic loading was applied to investigate the influence of structural and mechanical stimulation on cell arrangement and proliferation. The results demonstrated that the 75° scaffold exhibited auxetic characteristics in all compression directions and possessed anti-fracture properties. The 75° scaffold also promoted cell proliferation by structural design. Cyclic compression facilitated the nuclear translocation of YAP, further enhancing cell growth. The combination of anti-fracture properties and the promotion of cell proliferation makes auxetic structures highly promising for extensive applications.
Collapse
Affiliation(s)
- Xuezheng Geng
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yan Yao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China; School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Huiwen Huang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qiao Li
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China; School of Engineering Medicine, Beihang University, Beijing 100191, China; State Key Laboratory of Virtual Reality Technology and Systems, Beihang University, Beijing 100191, China.
| | - Lizhen Wang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China; State Key Laboratory of Virtual Reality Technology and Systems, Beihang University, Beijing 100191, China
| | - Yubo Fan
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115 Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing 100191, China; School of Engineering Medicine, Beihang University, Beijing 100191, China; State Key Laboratory of Virtual Reality Technology and Systems, Beihang University, Beijing 100191, China.
| |
Collapse
|
3
|
Shirali D, Emadi R, Khodaei M, Emadi H, Arab Eshagh Abadi M, Tayebi L. Surface modification of 3D-printed polylactic acid-hardystonite scaffold for bone tissue engineering. Int J Biol Macromol 2025; 308:142496. [PMID: 40139588 DOI: 10.1016/j.ijbiomac.2025.142496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/10/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
This study aims to synthesize hardystonite bioceramic powder using the sol-gel method and fabricate composite scaffolds of poly lactic acid (PLA)-hardystonite (Har) with different ceramic contents (0, 10, 20, and 30 wt%) through melt-based 3D-printing. X-ray diffraction (XRD) analysis of the ceramic powder and composites indicated that the characteristic peaks matched well with peak intensities increasing proportionally with the ceramic content in the composites. Porosity measurement using the Archimedes method revealed an increase in pore size and overall porosity with increasing Har content of the composite. Compression testing on 3D-printed composite scaffolds demonstrated that the compressive strength of the scaffolds increased from 25.9 ± 0.1 MPa for pure PLA scaffolds to 34.5 ± 2.1 MPa for PLA-20Har (scaffolds containing 20 wt% Har), while PLA-30Har scaffolds exhibited a decrease in compressive strength of approximately 10 %. Tensile testing on rigid 3D-printed composite samples indicated optimal mechanical properties for PLA-30Har composite. Additionally, the water contact angle decreased with higher ceramic content, from 71.9 ± 4.1° for pure PLA to 57.9 ± 3.1° for PLA-30Har, indicating improved wettability. The scaffolds exhibited favorable biocompatibility, biodegradability, and enhanced cell adhesion with higher ceramic content. Surface modification of the optimal scaffold (PLA-20Har) significantly reduced the water contact angle and further improved cell adhesion. These results suggest that these biocompatible scaffolds have strong potential for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Danial Shirali
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Rahmatollah Emadi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Mohammad Khodaei
- Materials Engineering Group, Golpayegan College of Engineering, Isfahan University of Technology, Golpayegan 87717-67498, Iran.
| | - Hosein Emadi
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran 14176-14411, Iran
| | | | - Lobat Tayebi
- Institute for Engineering in Medicine, Health, & Human Performance (EnMed), Batten College of Engineering and Technology, Old Dominion University, Norfolk, VA, 23529, USA
| |
Collapse
|
4
|
Topçu İnce B, Guieu S, Timur SS, Reçber T, Nemutlu E, Vaz Fernandes MH, Eroğlu H. Design and characterization of memantine and donepezil loaded 3D scaffolds. Pharm Dev Technol 2025; 30:488-504. [PMID: 40237315 DOI: 10.1080/10837450.2025.2493256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
Memantine HCl (MEM) and Donepezil HCl (DON) are widely used separately and in combination to treat Alzheimer's disease, and some studies suggest that these drugs may also prevent bone fractures and promote bone regeneration. For this purpose, we formulated fiber-based 3D scaffolds for local delivery of MEM/DON to improve the regeneration process of bone fractures. First, Poly (ε-caprolactone) (PCL)-based MEM/DON-loaded nanofibrous membranes were produced by electrospinning, and then these nanofibrous membranes were transformed into 3D scaffolds using the thermally induced self-agglomeration (TISA) method. Encapsulation efficiency after these two steps was found to be around 20%. Analyses confirmed that the 3D scaffolds have a morphology similar to the extracellular matrix, and that their hydrophilicity, swelling ratio, porosity, and degradation rate were adequate for bone tissue regeneration. Release studies show that the scaffolds provide an initial burst release of the drugs, followed by a sustained release for 21 days. These 3D scaffolds did not show any cytotoxic effect on the L-929 cell line, and increased cell viability over time indicates that they can be used in tissue engineering applications.
Collapse
Affiliation(s)
- Betül Topçu İnce
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Hacettepe University, Ankara, Turkey
| | - Samuel Guieu
- CICECO-Aveiro Institute of Materials, Department of Materials and Ceramic Engineering, University of Aveiro, Aveiro, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Selin Seda Timur
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Hacettepe University, Ankara, Turkey
| | - Tuba Reçber
- Faculty of Pharmacy, Department of Analytical Chemistry, Hacettepe University, Ankara, Turkey
| | - Emirhan Nemutlu
- Faculty of Pharmacy, Department of Analytical Chemistry, Hacettepe University, Ankara, Turkey
| | - Maria Helena Vaz Fernandes
- CICECO-Aveiro Institute of Materials, Department of Materials and Ceramic Engineering, University of Aveiro, Aveiro, Portugal
| | - Hakan Eroğlu
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
5
|
Xu T, Rao J, Mo Y, Lam ACH, Yang Y, Wong SWF, Wong KH, Zhao X. 3D printing in musculoskeletal interface engineering: Current progress and future directions. Adv Drug Deliv Rev 2025; 219:115552. [PMID: 40032068 DOI: 10.1016/j.addr.2025.115552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/17/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
The musculoskeletal system relies on critical tissue interfaces for its function; however, these interfaces are often compromised by injuries and diseases. Restoration of these interfaces is complex by nature which renders traditional treatments inadequate. An emerging solution is three-dimensional printing, which allows for precise fabrication of biomimetic scaffolds to enhance tissue regeneration. This review summarizes the use of 3D printing in creating scaffolds for musculoskeletal interfaces, mainly focusing on advanced techniques such as multi-material printing, bioprinting, and 4D printing. We emphasize the significance of mimicking natural tissue gradients and the selection of appropriate biomaterials to ensure scaffold success. The review outlines state-of-the-art 3D printing technologies, varying from extrusion, inkjet and laser-assisted bioprinting, which are crucial for producing scaffolds with tailored mechanical and biological properties. Applications in cartilage-bone, intervertebral disc, tendon/ligament-bone, and muscle-tendon junction engineering are discussed, highlighting the potential for improved integration and functionality. Furthermore, we address challenges in material development, printing resolution, and the in vivo performance of scaffolds, as well as the prospects for clinical translation. The review concludes by underscoring the transformative potential of 3D printing to advance orthopedic medicine, offering a roadmap for future research at the intersection of biomaterials, drug delivery, and tissue engineering.
Collapse
Affiliation(s)
- Tianpeng Xu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Jingdong Rao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Yongyi Mo
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Avery Chik-Him Lam
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Yuhe Yang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region; The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Sidney Wing-Fai Wong
- Industrial Centre, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Ka-Hing Wong
- Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region; The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Research Institute for Intelligent Wearable Systems, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
6
|
Ye S, Cao Q, Ni P, Xiong S, Zhong M, Yuan T, Shan J, Liang J, Fan Y, Zhang X. A ceramic microbridge microfluidic chip to study osteogenic differentiation of mesenchymal stem cells in bioactive ceramic immune microenvironment. Bioact Mater 2025; 45:520-533. [PMID: 39735335 PMCID: PMC11681893 DOI: 10.1016/j.bioactmat.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/13/2024] [Accepted: 11/05/2024] [Indexed: 12/31/2024] Open
Abstract
Bioactive ceramics have been used in bone tissue repair and regeneration. However, because of the complex in vivo osteogenesis process, long cycle, and difficulty of accurately tracking, the mechanism of interaction between materials and cells has yet to be fully understood, hindering its development. The ceramic microbridge microfluidic chip system may solve the problem and provide an in vitro method to simulate the microenvironment in vivo. Nevertheless, the complex microenvironment parameters of the chip system need to be studied in detail. Computer simulation bionics can provide clues for the setting of microenvironment parameters. This study used a computational bionic model to simulate the bone growth process in the presence of immune-related factors. The osteoblast differentiation of mesenchymal stem cells of calcium phosphate ceramics in a macrophage-dominated immune microenvironment was studied using a microfluidic chip system. The computational biomimetic model and microfluidic chip findings were basically consistent with the reported results of the animal experiments. These findings suggest that studying the osteogenic behavior of calcium phosphate ceramics using a microfluidic chip model is feasible. The method model provided in this study can be extended to other biomaterials, providing a viable path for their research and evaluation.
Collapse
Affiliation(s)
- Sheng Ye
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
- School of Big Health & Intelligent Engineering, Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Nuclear Industry 416 Hospital, the 2nd Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610057, China
| | - Quanle Cao
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Panxianzhi Ni
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Shuting Xiong
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Meng Zhong
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Tun Yuan
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
- Sichuan Testing Centre for Biomaterials and Medical Devices, Chengdu, Sichuan, 610064, China
| | - Jing Shan
- Department of Gastroenterology, the 3rd People's Hospital of Chengdu, Southwest Jiaotong University, Chengdu, Sichuan, 610064, China
| | - Jie Liang
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
- Sichuan Testing Centre for Biomaterials and Medical Devices, Chengdu, Sichuan, 610064, China
| | - Yujiang Fan
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Xingdong Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| |
Collapse
|
7
|
Merle M, Lagarrigue P, Wang S, Duployer B, Tenailleau C, Müller WEG, Poquillon D, Combes C, Soulié J. Freeze-Cast Composites of Alginate/Pyrophosphate-Stabilized Amorphous Calcium Carbonate: From the Nanoscale Structuration to the Macroscopic Properties. ACS Biomater Sci Eng 2025; 11:1198-1211. [PMID: 39772404 DOI: 10.1021/acsbiomaterials.4c01396] [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: 01/11/2025]
Abstract
Pyrophosphate-stabilized amorphous calcium carbonates (PyACC) are promising compounds for bone repair due to their ability to release calcium, carbonate, and phosphate ions following pyrophosphate hydrolysis. However, shaping these metastable and brittle materials using conventional methods remains a challenge, especially in the form of macroporous scaffolds, yet essential to promote cell colonization. To overcome these limitations, this article describes for the first time the design and multiscale characterization of freeze-cast alginate (Alg)-PyACC nanocomposite scaffolds. The study initially focused on the synthesis of Alg-PyACC powder through in situ coprecipitation. The presence of alginate chains in the vicinity of the PyACC was shown to affect both the powder reactivity and the release of calcium ions when placed in water (XRD, chemical titrations). In vitro cellular assays confirmed the biocompatibility of Alg-PyACC powder, supporting its use as a filler in scaffolds for bone substitutes. In a second step, the freeze-casting process was carried out using these precursor powders with varying rates of inorganic fillers. The resulting scaffolds were compared in terms of pore size and gradient (via SEM, X-ray microtomography, and mercury intrusion porosimetry). All scaffolds exhibited a pore size gradient oriented along the solidification axis, featuring unidirectional, lamellar, and interconnected pores. Interestingly, we found that the pore size and wall thickness could be controlled by the filler rate. This effect was attributed to the in situ cross-linking of alginate chains by released Ca2+ ions from the fillers, which increased viscosity, affecting temperature-driven segregation during the freezing step. Different multiscale organizations of the porosity and spatial distribution of fillers (FEG-SEM) were correlated with changes in the scaffold mechanical properties (tested via uniaxial compression). With such tunable porous and mechanical properties, Alg-PyACC composite scaffolds present attractive opportunities for specific bone substitute applications.
Collapse
Affiliation(s)
- Marion Merle
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, ENSIACET, 4 allée Emile Monso, Toulouse 31030, France
| | - Prescillia Lagarrigue
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, ENSIACET, 4 allée Emile Monso, Toulouse 31030, France
| | - Shunfeng Wang
- Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz 55128, Germany
| | - Benjamin Duployer
- CIRIMAT, Université Toulouse 3 Paul Sabatier, Toulouse INP, CNRS, Université de Toulouse, 118 Route de Narbonne, Toulouse 31062, France
| | - Christophe Tenailleau
- CIRIMAT, Université Toulouse 3 Paul Sabatier, Toulouse INP, CNRS, Université de Toulouse, 118 Route de Narbonne, Toulouse 31062, France
| | - Werner E G Müller
- Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz 55128, Germany
| | - Dominique Poquillon
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, ENSIACET, 4 allée Emile Monso, Toulouse 31030, France
| | - Christèle Combes
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, ENSIACET, 4 allée Emile Monso, Toulouse 31030, France
| | - Jérémy Soulié
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, ENSIACET, 4 allée Emile Monso, Toulouse 31030, France
| |
Collapse
|
8
|
Wang C, Liu A, Zhao Z, Ying T, Deng S, Jian Z, Zhang X, Yi C, Li D. Application and progress of 3D printed biomaterials in osteoporosis. Front Bioeng Biotechnol 2025; 13:1541746. [PMID: 39968010 PMCID: PMC11832546 DOI: 10.3389/fbioe.2025.1541746] [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: 12/08/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Osteoporosis results from a disruption in skeletal homeostasis caused by an imbalance between bone resorption and bone formation. Conventional treatments, such as pharmaceutical drugs and hormone replacement therapy, often yield suboptimal results and are frequently associated with side effects. Recently, biomaterial-based approaches have gained attention as promising alternatives for managing osteoporosis. This review summarizes the current advancements in 3D-printed biomaterials designed for osteoporosis treatment. The benefits of biomaterial-based approaches compared to traditional systemic drug therapies are discussed. These 3D-printed materials can be broadly categorized based on their functionalities, including promoting osteogenesis, reducing inflammation, exhibiting antioxidant properties, and inhibiting osteoclast activity. 3D printing has the advantages of speed, precision, personalization, etc. It is able to satisfy the requirements of irregular geometry, differentiated composition, and multilayered structure of articular osteochondral scaffolds with boundary layer structure. The limitations of existing biomaterials are critically analyzed and future directions for biomaterial-based therapies are considered.
Collapse
Affiliation(s)
- Chenxu Wang
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Aiguo Liu
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Ziwen Zhao
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Ting Ying
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Shuang Deng
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Zhen Jian
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xu Zhang
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Chengqing Yi
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Dejian Li
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
9
|
Syamal S, Taritsa IC, Alvarez AH, Schuster K, Foppiani J, Kaplan D, Lin SJ. Evaluating the Mechanical Strength of 3-Dimensionally Printed Implants in Septorhinoplasty through Finite Element Analysis. Plast Reconstr Surg 2025; 155:319e-333e. [PMID: 38923894 DOI: 10.1097/prs.0000000000011600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
BACKGROUND Autologous nasoseptal cartilage grafts are used to correct nasal asymmetry and deviation in rhinoplasty, but patients who have undergone multiple operations may have limited autologous cartilage tissue available. L-strut implants created on a 3-dimensional (3D) printer may address these challenges in the future, but their mechanical strength is understudied. Silk fibroin-gelatin (SFG), polycaprolactone (PCL), and polylactide (PLA) are bioinks known for their strength. The authors present finite element analysis (FEA) models comparing the mechanical strength of 3D-printed SFG, PCL, and PLA implants with nasoseptal cartilage grafts when autologous or allografts are not available. METHODS FEA models compared the stress and deformation responses of 3D-printed solid and scaffold implant replacements to cartilage. To simulate a daily force from overlying soft tissue, a unidirectional load was applied at the "keystone" region given its structural role and compared with native cartilaginous properties. RESULTS The 3D-printed solid SFG, PCL, and PLA and scaffold PCL and PLA models demonstrated lower deformations compared with cartilage. Solid SFG balanced strength and flexibility. The maximum stress was below all materials' yield stresses, suggesting that their deformations are unlikely permanent under a daily load. CONCLUSIONS The authors' FEA models suggest that 3D-printed L-strut implants carry promising mechanical strength. Solid SFG results mimicked cartilage's mechanical behavior. Thus, scaffold SFG merits further geometric optimization for potential use for cartilage substitution. The 3D-printed septal cartilage replacement implants can potentially enhance surgical management of patients who lack available donor cartilage in select settings. CLINICAL RELEVANCE STATEMENT Computational simulations can evaluate the strength of 3D-printed implants and their potential to replace septal cartilage in septorhinoplasty. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
Affiliation(s)
- Sujata Syamal
- From the Division of Continuing Education, Harvard University Extension School Cambridge
| | - Iulianna C Taritsa
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Angelica Hernandez Alvarez
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Kirsten Schuster
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Jose Foppiani
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School
| | - David Kaplan
- Department of Biomedical Engineering, Tufts University
| | - Samuel J Lin
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School
- Department of Biomedical Engineering, Tufts University
| |
Collapse
|
10
|
Zhang Z, Shang W, Lin L. Hydroxyapatite Chitosan Gradient Pore Scaffold Activates Oxidative Phosphorylation Pathway to Induce Bone Formation. FRONT BIOSCI-LANDMRK 2025; 30:26299. [PMID: 39862088 DOI: 10.31083/fbl26299] [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: 08/27/2024] [Revised: 11/15/2024] [Accepted: 11/29/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND In this study, we prepared a porous gradient scaffold with hydroxyapatite microtubules (HAMT) and chitosan (CHS) and investigated osteogenesis induced by these scaffolds. METHODS The arrangement of wax balls in the mold can control the size and distribution of the pores of the scaffold, and form an interconnected gradient pore structure. The scaffolds were systematically evaluated in vitro and in vivo for biocompatibility, biological activity, and regulatory mechanisms. RESULTS The porosity of the four scaffolds was more than 80%. The 50% and 70% HAMT-CHS scaffolds formed an excellent gradient pore structure, with interconnected pores. Furthermore, the 70% HAMT-CHS scaffold showed better anti-compressive deformation ability. In vitro experiments indicated that the scaffolds had good biocompatibility, promoted the expression of osteogenesis-related genes and proteins, and activated the oxidative phosphorylation pathway to promote bone regeneration. Eight weeks after implanting the HAMT-CHS scaffold in rat skull defects, new bone formation was observed in vivo by micro-computed tomographic (CT) staining. The obtained data were statistically analyzed, and the p-value < 0.05 was statistically significant. CONCLUSION HAMT-CHS scaffolds can accelerate osteogenesis in bone defects, potentially through the activation of the oxidative phosphorylation pathway. These results highlight the potential therapeutic application of HAMT-CHS scaffolds.
Collapse
Affiliation(s)
- Zeliang Zhang
- Department of Stomatology, The First Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, China
| | - Wei Shang
- Department of Stomatology, The Affiliated Heping Hospital of Changzhi Medical College, 046000 Changzhi, Shanxi, China
| | - Lisong Lin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fujian Provincial Key Laboratory of Stomatology, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, 350005 Fuzhou, Fujian, China
| |
Collapse
|
11
|
Chen X, Zhang X, Sun J, Zhang R, Liang X, Long J, Yao J, Chen X, Wang H, Zhang Y, Leng J, Lu R. Near-Field Direct Writing Based on Piezoelectric Micromotion for the Programmable Manufacturing of Serpentine Structures. MICROMACHINES 2024; 15:1478. [PMID: 39770231 PMCID: PMC11727834 DOI: 10.3390/mi15121478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Serpentine microstructures offer excellent physical properties, making them highly promising in applications in stretchable electronics and tissue engineering. However, existing fabrication methods, such as electrospinning and lithography, face significant challenges in producing microscale serpentine structures that are cost-effective, efficient, and controllable. These methods often struggle with achieving precise control over fiber morphology and scalability. In this study, we developed a near-field direct writing (NFDW) technique incorporating piezoelectric micromotion to enable the precise fabrication of serpentine micro-/nanofibers by incorporating micromotion control with macroscopic movement. Modifying the fiber structure allowed for adjustments to the mechanical properties, including tunable extensibility and distinct characteristics. Through the control of the frequency and amplitude of the piezoelectric signal, the printing errors were reduced to below 9.48% in the cycle length direction and 6.33% in the peak height direction. A predictive model for the geometrical extensibility of serpentine structures was derived from Legendre's incomplete elliptic integral of the second kind and incorporated an error correction factor, which significantly reduced the calculation errors in predicting geometric elongation, by 95.85%. The relationship between microstructure bending and biomimetic non-linear mechanical behavior was explored through tensile testing. By controlling the input electrical signals, highly ordered serpentine microstructures were successfully fabricated, demonstrating potential for use in biomimetic mechanical scaffolds.
Collapse
Affiliation(s)
- Xun Chen
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Xuanzhi Zhang
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Jianfeng Sun
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Rongguang Zhang
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Xuanyang Liang
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Jiecai Long
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Jingsong Yao
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Xin Chen
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Han Wang
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Yu Zhang
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Jiewu Leng
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China; (X.C.); (X.Z.); (J.S.); (R.Z.); (J.Y.); (X.C.); (H.W.); (Y.Z.); (J.L.)
- School of Electromechnical Engineering, Guangdong University of Technology, Guangzhou 510006, China;
| | - Renquan Lu
- Guangdong Provincial Key Laboratory of Intelligent Decision and Cooperative Control, School of Automation, Guangdong University of Technology, Guangzhou 510006, China;
| |
Collapse
|
12
|
Tang X, Zhou F, Wang S, Wang G, Bai L, Su J. Bioinspired injectable hydrogels for bone regeneration. J Adv Res 2024:S2090-1232(24)00486-7. [PMID: 39505143 DOI: 10.1016/j.jare.2024.10.032] [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: 01/07/2024] [Revised: 09/28/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024] Open
Abstract
The effective regeneration of bone/cartilage defects remains a significant clinical challenge, causing irreversible damage to millions annually.Conventional therapies such as autologous or artificial bone grafting often yield unsatisfactory outcomes, emphasizing the urgent need for innovative treatment methods. Biomaterial-based strategies, including hydrogels and active scaffolds, have shown potential in promoting bone/cartilage regeneration. Among them, injectable hydrogels have garnered substantial attention in recent years on account of their minimal invasiveness, shape adaptation, and controlled spatiotemporal release. This review systematically discusses the synthesis of injectable hydrogels, bioinspired approaches-covering microenvironment, structural, compositional, and bioactive component-inspired strategies-and their applications in various bone/cartilage disease models, highlighting bone/cartilage regeneration from an innovative perspective of bioinspired design. Taken together, bioinspired injectable hydrogels offer promising and feasible solutions for promoting bone/cartilage regeneration, ultimately laying the foundations for clinical applications. Furthermore, insights into further prospective directions for AI in injectable hydrogels screening and organoid construction are provided.
Collapse
Affiliation(s)
- Xuan Tang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an 710000, China
| | - Sicheng Wang
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Zhongye Hospital, Shanghai 201900, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Wenzhou Institute of Shanghai University, Wenzhou 325000, China.
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
13
|
Liu Z, Jia J, Lei Q, Wei Y, Hu Y, Lian X, Zhao L, Xie X, Bai H, He X, Si L, Livermore C, Kuang R, Zhang Y, Wang J, Yu Z, Ma X, Huang D. Electrohydrodynamic Direct-Writing Micro/Nanofibrous Architectures: Principle, Materials, and Biomedical Applications. Adv Healthc Mater 2024; 13:e2400930. [PMID: 38847291 DOI: 10.1002/adhm.202400930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/21/2024] [Indexed: 07/05/2024]
Abstract
Electrohydrodynamic (EHD) direct-writing has recently gained attention as a highly promising additive manufacturing strategy for fabricating intricate micro/nanoscale architectures. This technique is particularly well-suited for mimicking the extracellular matrix (ECM) present in biological tissue, which serves a vital function in facilitating cell colonization, migration, and growth. The integration of EHD direct-writing with other techniques has been employed to enhance the biological performance of scaffolds, and significant advancements have been made in the development of tailored scaffold architectures and constituents to meet the specific requirements of various biomedical applications. Here, a comprehensive overview of EHD direct-writing is provided, including its underlying principles, demonstrated materials systems, and biomedical applications. A brief chronology of EHD direct-writing is provided, along with an examination of the observed phenomena that occur during the printing process. The impact of biomaterial selection and architectural topographic cues on biological performance is also highlighted. Finally, the major limitations associated with EHD direct-writing are discussed.
Collapse
Affiliation(s)
- Zhengjiang Liu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
| | - Jinqiao Jia
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
| | - Qi Lei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Yan Wei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Yinchun Hu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Xiaojie Lian
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Liqin Zhao
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Xin Xie
- Xellar Biosystems, Cambridge, MA, 02458, USA
| | - Haiqing Bai
- Xellar Biosystems, Cambridge, MA, 02458, USA
| | - Xiaomin He
- Xellar Biosystems, Cambridge, MA, 02458, USA
| | - Longlong Si
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Carol Livermore
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Rong Kuang
- Zhejiang Institute for Food and Drug Control, Hangzhou, 310000, P. R. China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
| | - Jiucun Wang
- Human Phenome Institute, Fudan University, Shanghai, 200433, P. R. China
| | - Zhaoyan Yu
- Shandong Public Health Clinical Center, Shandong University, Jinan, 250000, P. R. China
| | - Xudong Ma
- Cytori Therapeutics LLC., Shanghai, 201802, P. R. China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| |
Collapse
|
14
|
Kirmanidou Y, Chatzinikolaidou M, Michalakis K, Tsouknidas A. Clinical translation of polycaprolactone-based tissue engineering scaffolds, fabricated via additive manufacturing: A review of their craniofacial applications. BIOMATERIALS ADVANCES 2024; 162:213902. [PMID: 38823255 DOI: 10.1016/j.bioadv.2024.213902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/17/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
The craniofacial region is characterized by its intricate bony anatomy and exposure to heightened functional forces presenting a unique challenge for reconstruction. Additive manufacturing has revolutionized the creation of customized scaffolds with interconnected pores and biomimetic microarchitecture, offering precise adaptation to various craniofacial defects. Within this domain, medical-grade poly(ε-caprolactone) (PCL) has been extensively used for the fabrication of 3D printed scaffolds, specifically tailored for bone regeneration. Its adoption for load-bearing applications was driven mainly by its mechanical properties, adjustable biodegradation rates, and high biocompatibility. The present review aims to consolidating current insights into the clinical translation of PCL-based constructs designed for bone regeneration. It encompasses recent advances in enhancing the mechanical properties and augmenting biodegradation rates of PCL and PCL-based composite scaffolds. Moreover, it delves into various strategies improving cell proliferation and the osteogenic potential of PCL-based materials. These strategies provide insight into the refinement of scaffold microarchitecture, composition, and surface treatments or coatings, that include certain bioactive molecules such as growth factors, proteins, and ceramic nanoparticles. The review critically examines published data on the clinical applications of PCL scaffolds in both extraoral and intraoral craniofacial reconstructions. These applications include cranioplasty, nasal and orbital floor reconstruction, maxillofacial reconstruction, and intraoral bone regeneration. Patient demographics, surgical procedures, follow-up periods, complications and failures are thoroughly discussed. Although results from extraoral applications in the craniofacial region are encouraging, intraoral applications present a high frequency of complications and related failures. Moving forward, future studies should prioritize refining the clinical performance, particularly in the domain of intraoral applications, and providing comprehensive data on the long-term outcomes of PCL-based scaffolds in bone regeneration. Future perspective and limitations regarding the transition of such constructs from bench to bedside are also discussed.
Collapse
Affiliation(s)
- Y Kirmanidou
- Laboratory for Biomaterials and Computational Mechanics, Department of Mechanical Engineering, University of Western Macedonia, University Campus ZEP, 50100 Kozani, Greece
| | - M Chatzinikolaidou
- Department of Materials Science and Engineering, University of Crete, 70013 Heraklion, Greece; Foundation for Research and Technology Hellas (FO.R.T.H), Institute of Electronic Structure and Laser (IESL), 70013 Heraklion, Greece
| | - K Michalakis
- Laboratory of Biomechanics, Department of Restorative Sciences & Biomaterials, Henry M. Goldman School of Dental Medicine, Boston University, Boston MA-02111, USA; Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, USA
| | - A Tsouknidas
- Laboratory for Biomaterials and Computational Mechanics, Department of Mechanical Engineering, University of Western Macedonia, University Campus ZEP, 50100 Kozani, Greece; Laboratory of Biomechanics, Department of Restorative Sciences & Biomaterials, Henry M. Goldman School of Dental Medicine, Boston University, Boston MA-02111, USA.
| |
Collapse
|
15
|
Zhang X, Zhu S, Liang Y, Jiang H, Cui Z, Li Z. Poloxamer 407 modified collagen/β-tricalcium phosphate scaffold for localized delivery of alendronate. J Biomater Appl 2024; 39:179-194. [PMID: 38842552 DOI: 10.1177/08853282241257613] [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: 06/07/2024]
Abstract
Systemic administration of alendronate is associated with various adverse reactions in clinical settings. To mitigate these side effects, poloxamer 407 (P-407) modified with cellulose was chosen to encapsulate alendronate. This drug-loaded system was then incorporated into a collagen/β-tricalcium phosphate (β-TCP) scaffold to create a localized drug delivery system. Nuclear magnetic resonance spectrum and rheological studies revealed hydrogen bonding between P-407 and cellulose as well as a competitive interaction with water that contributed to the delayed release of alendronate (ALN). Analysis of the degradation kinetics of P-407 and release kinetics of ALN indicated zero-order kinetics for the former and Fickian or quasi-Fickian diffusion for the latter. The addition of cellulose, particularly carboxymethyl cellulose (CMC), inhibited the degradation of P-407 and prolonged the release of ALN. The scaffold's structure increased the contact area of P-407 with the PBS buffer, thereby, influencing the release rate of ALN. Finally, biocompatibility testing demonstrated that the drug delivery system exhibited favorable cytocompatibility and hemocompatibility. Collectively, these findings suggest that the drug delivery system holds promise for implantation and bone healing applications.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, China
| | - Shengli Zhu
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, China
| | - Yanqin Liang
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, China
| | - Hui Jiang
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, China
| | - Zhenduo Cui
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, China
| | - Zhaoyang Li
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, China
| |
Collapse
|
16
|
Chen X, Liu W, Su C, Shan J, Li X, Chai Y, Yu Y, Wen G. Multimodal effects of an extracellular matrix on cellular morphology, dynamics and functionality. J Mater Chem B 2024; 12:7946-7958. [PMID: 39041314 DOI: 10.1039/d4tb00360h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Articular cartilage defects can lead to pain and even disability in patients and have significant socioeconomic loss. Repairing articular cartilage defects remains a long-term challenge in medicine owing to the limited ability of cartilage to regenerate. At present, the treatment methods adopted in clinical practice have many limitations, thereby necessitating the rapid development of biomaterials. Among them, decellularized biomaterials have been particularly prominent, with numerous breakthroughs in research progress and translational applications. Although many studies show that decellularized cartilage biomaterials promote tissue regeneration, any differences in cellular morphology, dynamics, and functionality among various biomaterials upon comparison have not been reported. In this study, we prepared cartilage-derived extracellular matrix (cdECM) biomaterials with different bioactive contents and various physical properties to compare their effects on the morphology, dynamics and functionality of chondrocytes. This cellular multimodal analysis of the characteristics of cdECM biomaterials provided a theoretical basis for understanding the interactions between biomaterials and cells, thus laying an experimental foundation for the translation and application of decellularized cartilage biomaterials in the treatment of cartilage defects.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Wenhao Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Chi Su
- Deyang Hospital of Integrated Traditional Chinese and Western Medicine, Sichuan, 618000, China
| | - Jianyang Shan
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Xiang Li
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Yaling Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Gen Wen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| |
Collapse
|
17
|
Mondal P, Chatterjee K. Multibiofunctional Self-healing Adhesive Injectable Nanocomposite Polysaccharide Hydrogel. Biomacromolecules 2024; 25:4762-4779. [PMID: 38989826 DOI: 10.1021/acs.biomac.4c00016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Injectable hydrogels with good antimicrobial and antioxidant properties, self-healing characteristics, suitable mechanical properties, and therapeutic effects have great practical significance for developing treatments for pressing healthcare challenges. Herein, we have designed a novel, self-healing injectable hydrogel composite incorporating cross-linked biofunctional nanomaterials by mixing alginate aldehyde (Ox-Alg), quaternized chitosan (QCS), adipic acid dihydrazide (ADH), and copper oxide nanosheets surface functionalized with folic acid as the bioligand (F-CuO). Gelation was achieved under physiological conditions via the dynamic Schiff base cross-linking mechanism. The developed nanocomposite injectable hydrogel demonstrated the fast self-healing ability essential to bear deformation and outstanding antibacterial properties along with ROS scavenging ability. Furthermore, the optimized formulation of our F-CuO-embedded injectable hydrogel exhibited excellent cytocompatibility, blood compatibility, and in vitro wound healing performance. Taken together, the F-CuO nanosheet cross-linked injectable hydrogel composite presented herein offers a promising candidate biomaterial with multifunctional properties to develop solutions for addressing clinical challenges.
Collapse
Affiliation(s)
- Pritiranjan Mondal
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India
| |
Collapse
|
18
|
Mi CH, Qi XY, Zhou YW, Ding YW, Wei DX, Wang Y. Advances in medical polyesters for vascular tissue engineering. DISCOVER NANO 2024; 19:125. [PMID: 39115796 PMCID: PMC11310390 DOI: 10.1186/s11671-024-04073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/25/2024] [Indexed: 08/11/2024]
Abstract
Blood vessels are highly dynamic and complex structures with a variety of physiological functions, including the transport of oxygen, nutrients, and metabolic wastes. Their normal functioning involves the close and coordinated cooperation of a variety of cells. However, adverse internal and external environmental factors can lead to vascular damage and the induction of various vascular diseases, including atherosclerosis and thrombosis. This can have serious consequences for patients, and there is an urgent need for innovative techniques to repair damaged blood vessels. Polyesters have been extensively researched and used in the treatment of vascular disease and repair of blood vessels due to their excellent mechanical properties, adjustable biodegradation time, and excellent biocompatibility. Given the high complexity of vascular tissues, it is still challenging to optimize the utilization of polyesters for repairing damaged blood vessels. Nevertheless, they have considerable potential for vascular tissue engineering in a range of applications. This summary reviews the physicochemical properties of polyhydroxyalkanoate (PHA), polycaprolactone (PCL), poly-lactic acid (PLA), and poly(lactide-co-glycolide) (PLGA), focusing on their unique applications in vascular tissue engineering. Polyesters can be prepared not only as 3D scaffolds to repair damage as an alternative to vascular grafts, but also in various forms such as microspheres, fibrous membranes, and nanoparticles to deliver drugs or bioactive ingredients to damaged vessels. Finally, it is anticipated that further developments in polyesters will occur in the near future, with the potential to facilitate the wider application of these materials in vascular tissue engineering.
Collapse
Affiliation(s)
- Chen-Hui Mi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Xin-Ya Qi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Yan-Wen Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Yan-Wen Ding
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Dai-Xu Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
- School of Clinical Medicine, Chengdu University, Chengdu, China.
- Shaanxi Key Laboratory for Carbon-Neutral Technology, Xi'an, 710069, China.
| | - Yong Wang
- Department of Interventional Radiology and Vascular Surgery, Second Affiliated Hospital of Hainan Medical University, Haikou, China.
| |
Collapse
|
19
|
Li J, Li X, Li X, Liang Z, Wang Z, Shahzad KA, Xu M, Tan F. Local Delivery of Dual Stem Cell-Derived Exosomes Using an Electrospun Nanofibrous Platform for the Treatment of Traumatic Brain Injury. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37497-37512. [PMID: 38980910 DOI: 10.1021/acsami.4c05004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Traumatic brain injury poses serious physical, psychosocial, and economic threats. Although systemic administration of stem cell-derived exosomes has recently been proven to be a promising modality for traumatic brain injury treatment, they come with distinct drawbacks. Luckily, various biomaterials have been developed to assist local delivery of exosomes to improve the targeting of organs, minimize nonspecific accumulation in vital organs, and ensure the protection and release of exosomes. In this study, we developed an electrospun nanofibrous scaffold to provide sustained delivery of dual exosomes derived from mesenchymal stem cells and neural stem cells for traumatic brain injury treatment. The electrospun nanofibrous scaffold employed a functionalized layer of polydopamine on electrospun poly(ε-caprolactone) nanofibers, thereby enhancing the efficient incorporation of exosomes through a synergistic interplay of adhesive forces, hydrogen bonding, and electrostatic interactions. First, the mesenchymal stem cell-derived exosomes and the neural stem cell-derived exosomes were found to modulate microglial polarization toward M2 phenotype, play an important role in the modulation of inflammatory responses, and augment axonal outgrowth and neural repair in PC12 cells. Second, the nanofibrous scaffold loaded with dual stem cell-derived exosomes (Duo-Exo@NF) accelerated functional recovery in a murine traumatic brain injury model, as it mitigated the presence of reactive astrocytes and microglia while elevating the levels of growth associated protein-43 and doublecortin. Additionally, multiomics analysis provided mechanistic insights into how dual stem cell-derived exosomes exerted its therapeutic effects. These findings collectively suggest that our novel Duo-Exo@NF system could function as an effective treatment modality for traumatic brain injury using sustained local delivery of dual exosomes from stem cells.
Collapse
Affiliation(s)
- Jiaojiao Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xuran Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai 200070, China
| | - Xiangyu Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai 200065, China
| | - Zhanping Liang
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai 200065, China
| | - Zhao Wang
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Khawar Ali Shahzad
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai 200070, China
| | - Maoxiang Xu
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai 200070, China
| | - Fei Tan
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai 200070, China
- The Royal College of Surgeons in Ireland, Dublin D02YN77, Ireland
- The Royal College of Surgeons of England, London WC2A3PE, U.K
| |
Collapse
|
20
|
Samatya Yilmaz S, Aytac A. Antibacterial wound dressing with cross-linked electrospun surface from reduced graphene oxide doped polyvinyl alcohol/sodium caseinate blends. Biopolymers 2024; 115:e23579. [PMID: 38578129 DOI: 10.1002/bip.23579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024]
Abstract
In this study, a new biomaterial with polyvinyl alcohol (PVA)/sodium caseinate (SodCa)/reduced graphene oxide (rGO) structure was developed. Antibacterial effective nanofibers were successfully produced by electrospinning method from 1%, 3%, 5%, and 7% rGO added PVA/SodCa (60:40, w:w) solution mixtures prepared for use as modern wound dressings. To create a usage area, especially in exuding wounds, hydrophilic PVA/SodCa/rGO electrospun mats were cross-linked by dipping them in a glutaraldehyde (GLA) bath. The surface micrographs of all nanofibers were homogeneous and smooth. rGO-doped biomaterials were obtained as thin nanofibers in the range of 301-348 nm. Nanofibers, which were completely soluble in water, after cross-linking preserved their existence in the range of 87%-81% at the end of the 24th hour in distilled water. It was reported that these biomaterials that persist in an aqueous environment show swelling behavior in the range of 275%-608%. The porosity of uncross-linked pure PVA/SodCa nanofibers increased by 46.75% after cross-linking. Moreover, the tensile strength of cross-linked PVA/SodCa electrospun mats increased in the presence of rGO. Provided that wound dressing is done every 24 h with 3% rGO-doped PVA/SodCa nanofiber and provided that wound dressing is done every 48 h with 5% rGO-doped PVA/SodCa nanofiber showed antibacterial activity against S. aureus as 99.38% and 99.55%, respectively.
Collapse
Affiliation(s)
| | - Ayse Aytac
- Chemical Engineering Department, Kocaeli University, Kocaeli, Turkey
- Polymer Science and Technology Department, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
21
|
Wu J, Jiao C, Yu H, Naqvi SMR, Ge M, Cai K, Liang H, Liu J, Zhao J, Tian Z, Wang D, Shen L. 3D printed barium titanate/calcium silicate composite biological scaffold combined with structural and material properties. BIOMATERIALS ADVANCES 2024; 158:213783. [PMID: 38295646 DOI: 10.1016/j.bioadv.2024.213783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 03/03/2024]
Abstract
In the case of a large bone defect, the human endogenous electrical field is no longer sufficient. Therefore, it is necessary to support structural electrical bone scaffolds. Barium titanate (BT) has received much attention in bone tissue engineering applications due to its biocompatibility and ability to maintain charged surfaces. However, its processability is poor and it does not have the biological activity to promote mineralization, which limits its use in bone repair. In this paper, a composite bone scaffold with excellent piezoelectric properties was prepared by combining 20 wt% calcium silicate. The influence of the light curing process on the properties of the piezoelectric biological scaffold was investigated by comparing it with the traditional piezoelectric ceramic molding method (dry pressing). Despite the structural features of 3D printing (layered structure, pore features), the piezoelectric and mechanical properties of the scaffold are weakened. However, 3D-printed scaffolds can combine structural and piezoelectric properties, which makes the 3D-printed scaffold more effective in terms of degradation and antibacterial performance. In terms of cell activity, piezoelectric properties attract proteins and nutrients into the scaffold, promoting cell growth and differentiation. Besides, it is undeniable that the pore structure of the scaffolds plays an important role in the biological properties. Finally, the 3D printed scaffolds have excellent antimicrobial properties due to the redox reaction under piezoelectric effect as well as structural characterization.
Collapse
Affiliation(s)
- Junnan Wu
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; Institute of Additive Manufacturing (3D Printing), Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Chen Jiao
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; Institute of Additive Manufacturing (3D Printing), Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Hanjiao Yu
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; Institute of Additive Manufacturing (3D Printing), Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Syed Mesum Raza Naqvi
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; Institute of Additive Manufacturing (3D Printing), Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Mengxing Ge
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; Institute of Additive Manufacturing (3D Printing), Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Kunzhan Cai
- Department of Dental Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Huixin Liang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jing Liu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Department of Stomatology, Nanjing 210000, China
| | - Jianfeng Zhao
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; Institute of Additive Manufacturing (3D Printing), Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Zongjun Tian
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; Institute of Additive Manufacturing (3D Printing), Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Dongsheng Wang
- College of Mechanical Engineering, Tongling University, Tongling 244061, China; Advanced Copper-based Materials Industry Generic Technology Research Center of Anhui Province, Tongling 244061, China.
| | - Lida Shen
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; Institute of Additive Manufacturing (3D Printing), Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China.
| |
Collapse
|
22
|
Shi J, Yao H, Wang B, Yang J, Liu D, Shang X, Chong H, Fei W, Wang DA. Construction of a Decellularized Multicomponent Extracellular Matrix Interpenetrating Network Scaffold by Gelatin Microporous Hydrogel 3D Cell Culture System. Macromol Rapid Commun 2024; 45:e2300508. [PMID: 38049086 DOI: 10.1002/marc.202300508] [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: 08/25/2023] [Revised: 11/25/2023] [Indexed: 12/06/2023]
Abstract
Interface tissue repair requires the construction of biomaterials with integrated structures of multiple protein types. Hydrogels that modulate internal porous structures provide a 3D microenvironment for encapsulated cells, making them promise for interface tissue repair. Currently, reduction of intrinsic immunogenicity and increase of bioactive extracellular matrix (ECM) secretion are issues to be considered in these materials. In this study, gelatin methacrylate (GelMA) hydrogel is used to encapsulate chondrocytes and construct a phase transition 3D cell culture system (PTCC) by utilizing the thermosensitivity of gelatin microspheres to create micropores within the hydrogel. The types of bioactive extracellular matrix protein formation by chondrocytes encapsulated in hydrogels are investigated in vitro. After 28 days of culture, GelMA PTCC forms an extracellular matrix predominantly composed of collagen type II, collagen type I, and fibronectin. After decellularization, the protein types and mechanical properties are well preserved, fabricating a decellularized tissue-engineered extracellular matrix and GelMA hydrogel interpenetrating network hydrogel (dECM-GelMA IPN) consisting of GelMA hydrogel as the first-level network and the ECM secreted by chondrocytes as the second-level network. This material has the potential to mediate the repair and regeneration of tendon-bone interface tissues with multiple protein types.
Collapse
Affiliation(s)
- Junli Shi
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Bowen Wang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Jian Yang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou, 225001, P. R. China
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Dianwei Liu
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou, 225001, P. R. China
| | - Xianfeng Shang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Hui Chong
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Wenyong Fei
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou, 225001, P. R. China
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong S.A.R
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, 518057, P. R. China
| |
Collapse
|
23
|
Norozi S, Ghollasi M, Salimi A, Halabian R, Shahrousvad M. Mesenchymal stem cells osteogenic differentiation by ZnO nanoparticles and polyurethane bimodal foam nanocomposites. Cell Tissue Bank 2024; 25:167-185. [PMID: 37103688 DOI: 10.1007/s10561-023-10090-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 04/06/2023] [Indexed: 04/28/2023]
Abstract
Mesenchymal stem cells with tissue repair capacity involve in regenerative medicine. MSCs can promote bone repair when employed with nano scaffolds/particles. Here, the MTT and Acridine Orange assay enabled the cytotoxic concentration of Zinc oxide nanoparticles and Polyurethane evaluation. Following culturing adipose tissue-derived MSCs, ADSCs' proliferation, growth, and osteogenic differentiation in the presence of PU with and without ZnO NPs is tracked by a series of biological assays, including Alkaline Phosphatase activity, Calcium deposition, alizarin red staining, RT-PCR, scanning electron microscope, and immunohistochemistry. The results showed boosted osteogenic differentiation of ADSCs in the presence of 1% PU scaffold and ZnO NPS and can thus apply as a new bone tissue engineering matrix. The expression level of Osteonectin, Osteocalcin, and Col1 increased in PU-ZnO 1% on the 7th and 14th days. There was an increase in the Runx2 gene expression on the 7th day of differentiation in PU-ZnO 1%, while it decreased on day 14th. In conclusion, Polyurethane nano scaffolds supported the MSCs' growth and rapid osteogenic differentiation. The PU-ZnO helps not only with cellular adhesion and proliferation but also with osteogenic differentiation.
Collapse
Affiliation(s)
- Shima Norozi
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mrazieh Ghollasi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mohsen Shahrousvad
- Caspian Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| |
Collapse
|
24
|
Rodríguez-Martín M, Aguilar JM, Castro-Criado D, Romero A. Characterization of Gelatin-Polycaprolactone Membranes by Electrospinning. Biomimetics (Basel) 2024; 9:70. [PMID: 38392116 PMCID: PMC10887028 DOI: 10.3390/biomimetics9020070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
New advances in materials science and medicine have enabled the development of new and increasingly sophisticated biomaterials. One of the most widely used biopolymers is polycaprolactone (PCL) because it has properties suitable for biomedical applications, tissue engineering scaffolds, or drug delivery systems. However, PCL scaffolds do not have adequate bioactivity, and therefore, alternatives have been studied, such as mixing PCL with bioactive polymers such as gelatin, to promote cell growth. Thus, this work will deal with the fabrication of nanofiber membranes by means of the electrospinning technique using PCL-based solutions (12 wt.% and 20 wt.%) and PCL with gelatin (12 wt.% and 8 wt.%, respectively). Formic acid and acetic acid, as well as mixtures of both in different proportions, have been used to prepare the preliminary solutions, thus supporting the electrospinning process by controlling the viscosity of the solutions and, therefore, the size and uniformity of the fibers. The physical properties of the solutions and the morphological, mechanical, and thermal properties of the membranes were evaluated. Results demonstrate that it is possible to achieve the determined properties of the samples with an appropriate selection of polymer concentrations as well as solvents.
Collapse
Affiliation(s)
- Manuel Rodríguez-Martín
- Department of Chemical Engineering, Faculty of Chemistry, University of Seville, 41012 Seville, Spain
| | - José Manuel Aguilar
- Department of Chemical Engineering, Faculty of Chemistry, University of Seville, 41012 Seville, Spain
| | - Daniel Castro-Criado
- Department of Chemical Engineering, Faculty of Chemistry, University of Seville, 41012 Seville, Spain
| | - Alberto Romero
- Department of Chemical Engineering, Faculty of Chemistry, University of Seville, 41012 Seville, Spain
| |
Collapse
|
25
|
De Mori A, Heyraud A, Tallia F, Blunn G, Jones JR, Roncada T, Cobb J, Al-Jabri T. Ovine Mesenchymal Stem Cell Chondrogenesis on a Novel 3D-Printed Hybrid Scaffold In Vitro. Bioengineering (Basel) 2024; 11:112. [PMID: 38391598 PMCID: PMC10886199 DOI: 10.3390/bioengineering11020112] [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: 11/21/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 02/24/2024] Open
Abstract
This study evaluated the use of silica/poly(tetrahydrofuran)/poly(ε-caprolactone) (SiO2/PTHF/PCL-diCOOH) 3D-printed scaffolds, with channel sizes of either 200 (SC-200) or 500 (SC-500) µm, as biomaterials to support the chondrogenesis of sheep bone marrow stem cells (oBMSC), under in vitro conditions. The objective was to validate the potential use of SiO2/PTHF/PCL-diCOOH for prospective in vivo ovine studies. The behaviour of oBMSC, with and without the use of exogenous growth factors, on SiO2/PTHF/PCL-diCOOH scaffolds was investigated by analysing cell attachment, viability, proliferation, morphology, expression of chondrogenic genes (RT-qPCR), deposition of aggrecan, collagen II, and collagen I (immunohistochemistry), and quantification of sulphated glycosaminoglycans (GAGs). The results showed that all the scaffolds supported cell attachment and proliferation with upregulation of chondrogenic markers and the deposition of a cartilage extracellular matrix (collagen II and aggrecan). Notably, SC-200 showed superior performance in terms of cartilage gene expression. These findings demonstrated that SiO2/PTHF/PCL-diCOOH with 200 µm pore size are optimal for promoting chondrogenic differentiation of oBMSC, even without the use of growth factors.
Collapse
Affiliation(s)
- Arianna De Mori
- School of Pharmacy and Biomedical Science, University of Portsmouth, St Micheal's Building, White Swan Road, Portsmouth PO1 2DT, UK
| | - Agathe Heyraud
- Department of Materials, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Francesca Tallia
- Department of Materials, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Gordon Blunn
- School of Pharmacy and Biomedical Science, University of Portsmouth, St Micheal's Building, White Swan Road, Portsmouth PO1 2DT, UK
| | - Julian R Jones
- Department of Materials, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Tosca Roncada
- Trinity Center for Biomedical Engineering, Trinity Biomedical Science Institute, Trinity College Dublin, 152-160 Pearse Street, DO2 R590 Dublin, Ireland
| | - Justin Cobb
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | - Talal Al-Jabri
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
26
|
Soltani L, Ghaneialvar H, Abbasi N, Bayat P, Nazari M. Chitosan/alginate scaffold enhanced with Berberis vulgaris extract for osteocyte differentiation of ovine fetal stem cells. Cell Biochem Funct 2024; 42:e3924. [PMID: 38269507 DOI: 10.1002/cbf.3924] [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: 10/14/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/26/2024]
Abstract
Designing biocompatible polymers using plant derivatives can be extremely useful in tissue engineering, nanomedicine, and many other fields of medicine. In this study, it was first looked into how chitosan/alginate scaffolds were made and characterized in the presence of berberine and barberry fruit extract. Second, the process of proliferation and differentiation of ovine fetal BM-MSCs (bone marrow-mesenchymal stem cells) was assessed on these scaffolds after BM-MSCs were extracted and confirmed by developing into osteocyte and adipose cells. To investigate the differentiation, treatment groups include (1) ovine fetal BM-MSCs were plated in Dulbecco's modified eagle medium culture medium with high glucose containing 10% fetal bovine serum and antibiotics (negative control), (2) ovine fetal BM-MSCs were plated in osteogenic differentiation medium (positive control group), (3) positive control group + barberry fruit extract, (4) positive control group + berberine, (5) ovine fetal BM-MSCs were plated in osteogenic differentiation medium on chitosan/alginate scaffold (hydrogel group), (6) ovine fetal BM-MSCs were plated in osteogenic differentiation medium on chitosan/alginate/barberry fruit extract scaffold (hydrogel group containing barberry fruit extract), and (7) ovine fetal BM-MSCs were plated in osteogenic differentiation medium on chitosan/alginate/berberine scaffold (hydrogel group containing berberine). Alkaline phosphatase (ALP) enzyme concentrations, mineralization rate using a calcium kit, and mineralization measurement by alizarin staining quantification were all found after 21 days of culture. In addition, real-time quantitative reverse transcription polymerase chain reaction was used to assess the expression of the ALP, COL1A2, and Runx2 genes. Days 5 and 7 had the lowest water absorption by the hydrogel scaffold containing barberry extract, which was significant in comparison to other groups (p < .05). Among the hydrogel scaffolds under study, the one containing barberry extract exhibited the lowest tensile strength, and this difference was statistically significant (p < .05). The chitosan/alginate hydrogel has the highest tensile strength of all of them. In comparison to the control and other treatment groups, the inclusion of berberine in the chitosan/alginate hydrogel significantly increased the expression of the ALP, Runx2, and COL1A2 genes (p < .05). The osteocyte differentiation of mesenchymal stem cells in in vitro settings appears to have been enhanced by the inclusion of berberine in the chitosan/alginate scaffold.
Collapse
Affiliation(s)
- Leila Soltani
- Department of Animal Sciences, Faculty of Agriculture, Razi University, Kermanshah, Iran
| | - Hori Ghaneialvar
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
- Department of Clinical Biochemistry, Medical School, Ilam University of Medical Sciences, Ilam, Iran
| | - Naser Abbasi
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
- Department of Pharmacology, Medical School, Ilam University of Medical Sciences, Ilam, Iran
| | - Parvaneh Bayat
- Department of Chemistry, Isfahan University of Technology, Ilam, Iran
| | - Maryam Nazari
- Applied Chemistry Department, Faculty of Chemistry, Razi University, Kermanshah, Iran
| |
Collapse
|
27
|
Javkhlan Z, Hsu SH, Chen RS, Chen MH. 3D-printed polycaprolactone scaffolds coated with beta tricalcium phosphate for bone regeneration. J Formos Med Assoc 2024; 123:71-77. [PMID: 37709573 DOI: 10.1016/j.jfma.2023.08.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND/PURPOSE 3D-printing technology is an important tool for the bone tissue engineering (BTE). The aim of this study was to investigate the interaction of polycaprolactone (PCL) scaffolds and modified mesh PCL coated with beta TCP (PCL/β-TCP) scaffolds with MG-63. METHODS This study used the fused deposition modeling (FDM) technique with the 3D printing technique to fabricate the thermoplastic polymer and composite scaffolds. Scaffold structure and coating quality were observed under a scanning electron microscope (SEM). MG-63 cells were injected and attached to the mesh-manufactured PCL scaffolds. The biocompatibility of mesh structured PCL and PCL/β-TCP scaffolds could be examined by measuring the viability of MG-63 cells of MTT assay. Bone cell differentiation was evaluated ALP activity by mineralization assay. RESULTS The results showed that both mesh PCL scaffolds and PCL/β-TCP scaffolds were non-toxic to the cells. The ALP activities of cells in PCL/β-TCP scaffolds groups were significant differences and better than PCL groups in all groups at all experimental dates. The mineralization process was time-dependent, and significantly higher mineralization of osteosarcoma cells was observed on PCL/β-TCP scaffolds at experimental dates. CONCLUSION We concluded that both meshes structured PCL and PCL/β-TCP scaffolds could promote the MG-63 cell growth, and PCL/β-TCP was better than the PCL scaffolds for the outcome of MG63 cell differentiation and mineralization.
Collapse
Affiliation(s)
- Zolzaya Javkhlan
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Sheng-Hao Hsu
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Rung-Shu Chen
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Min-Huey Chen
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
28
|
Long T, Xu T, Li R, Xu Z, Li D, Mu C, Yuan L, Mu Y. Emulsion template fabricated gelatin-based scaffold functionalized by dialdehyde starch complex with antibacterial antioxidant properties for accelerated wound healing. Int J Biol Macromol 2024; 254:127918. [PMID: 37977450 DOI: 10.1016/j.ijbiomac.2023.127918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/31/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
Gelatin and starch are considered as promising sustainable materials for their abundant production and good biodegradability. Efforts have been made to explore their medical application. Herein, scaffolds based on gelatin and starch with a preferred microstructure and antibacterial antioxidant property were fabricated by the emulsion template method. The dialdehyde starch was firstly combined with silver nanoparticles and curcumin to carry out the efficient hybrid antibacterial agent. Then, the gelatin microsphere of appropriate size was prepared by emulsification and gathered by the above agent to obtain gelatin-based scaffolds. The prepared scaffolds showed porous microstructures with high porosity of over 74 % and the preferred pore sizes of ∼65 μm, which is conducive to skin regeneration. Moreover, the scaffolds possessed a good swelling ability of over 640 %, good degradability of over 18 days, excellent blood compatibility, and cell compatibility. The promising antibacterial and antioxidant properties came from the hybrid antibacterial agent were affirmed. As expected, the gelatin-based scaffolds fabricated by the emulsion template method with a preferred microstructure can facilitate more adhered fibroblasts. In summary, gelatin-based scaffolds functionalized by starch-based complex expanded the application of abundant sustainable materials in the biomedical field, especially as antibacterial antioxidant wound dressings.
Collapse
Affiliation(s)
- Tao Long
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Ting Xu
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Rui Li
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Zhilang Xu
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Defu Li
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Changdao Mu
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Lun Yuan
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China; Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, PR China.
| | - Yandong Mu
- Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, PR China.
| |
Collapse
|
29
|
Chen P, Liao X. Kartogenin delivery systems for biomedical therapeutics and regenerative medicine. Drug Deliv 2023; 30:2254519. [PMID: 37665332 PMCID: PMC10478613 DOI: 10.1080/10717544.2023.2254519] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/14/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
Kartogenin, a small and heterocyclic molecule, has emerged as a promising therapeutic agent for incorporation into biomaterials, owing to its unique physicochemical and biological properties. It holds potential for the regeneration of cartilage-related tissues in various common conditions and injuries. Achieving sustained release of kartogenin through appropriate formulation and efficient delivery systems is crucial for modulating cell behavior and tissue function. This review provides an overview of cutting-edge kartogenin-functionalized biomaterials, with a primarily focus on their design, structure, functions, and applications in regenerative medicine. Initially, we discuss the physicochemical properties and biological functions of kartogenin, summarizing the underlying molecular mechanisms. Subsequently, we delve into recent advancements in nanoscale and macroscopic materials for the carriage and delivery of kartogenin. Lastly, we address the opportunities and challenges presented by current biomaterial developments and explore the prospects for their application in tissue regeneration. We aim to enhance the generation of insightful ideas for the development of kartogenin delivery materials in the field of biomedical therapeutics and regenerative medicine by providing a comprehensive understanding of common preparation methods.
Collapse
Affiliation(s)
- Peixing Chen
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, China
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, China
| | - Xiaoling Liao
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, China
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, China
| |
Collapse
|
30
|
Stafin K, Śliwa P, Piątkowski M. Towards Polycaprolactone-Based Scaffolds for Alveolar Bone Tissue Engineering: A Biomimetic Approach in a 3D Printing Technique. Int J Mol Sci 2023; 24:16180. [PMID: 38003368 PMCID: PMC10671727 DOI: 10.3390/ijms242216180] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
The alveolar bone is a unique type of bone, and the goal of bone tissue engineering (BTE) is to develop methods to facilitate its regeneration. Currently, an emerging trend involves the fabrication of polycaprolactone (PCL)-based scaffolds using a three-dimensional (3D) printing technique to enhance an osteoconductive architecture. These scaffolds are further modified with hydroxyapatite (HA), type I collagen (CGI), or chitosan (CS) to impart high osteoinductive potential. In conjunction with cell therapy, these scaffolds may serve as an appealing alternative to bone autografts. This review discusses research gaps in the designing of 3D-printed PCL-based scaffolds from a biomimetic perspective. The article begins with a systematic analysis of biological mineralisation (biomineralisation) and ossification to optimise the scaffold's structural, mechanical, degradation, and surface properties. This scaffold-designing strategy lays the groundwork for developing a research pathway that spans fundamental principles such as molecular dynamics (MD) simulations and fabrication techniques. Ultimately, this paves the way for systematic in vitro and in vivo studies, leading to potential clinical applications.
Collapse
Affiliation(s)
- Krzysztof Stafin
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, ul. Warszawska 24, PL 31-155 Kraków, Poland; (K.S.); (P.Ś.)
- Department of Biotechnology and Physical Chemistry, Faculty of Chemical Engineering and Technology, Cracow University of Technology, ul. Warszawska 24, PL 31-155 Kraków, Poland
| | - Paweł Śliwa
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, ul. Warszawska 24, PL 31-155 Kraków, Poland; (K.S.); (P.Ś.)
| | - Marek Piątkowski
- Department of Biotechnology and Physical Chemistry, Faculty of Chemical Engineering and Technology, Cracow University of Technology, ul. Warszawska 24, PL 31-155 Kraków, Poland
| |
Collapse
|
31
|
Crouch DJ, Sheridan CM, Behnsen JG, D’Sa RA, Bosworth LA. Cryo-Electrospinning Generates Highly Porous Fiber Scaffolds Which Improves Trabecular Meshwork Cell Infiltration. J Funct Biomater 2023; 14:490. [PMID: 37888155 PMCID: PMC10607045 DOI: 10.3390/jfb14100490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/15/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Human trabecular meshwork is a sieve-like tissue with large pores, which plays a vital role in aqueous humor outflow. Dysfunction of this tissue can occur, which leads to glaucoma and permanent vision loss. Replacement of trabecular meshwork with a tissue-engineered device is the ultimate objective. This study aimed to create a biomimetic structure of trabecular meshwork using electrospinning. Conventional electrospinning was compared to cryogenic electrospinning, the latter being an adaptation of conventional electrospinning whereby dry ice is incorporated in the fiber collector system. The dry ice causes ice crystals to form in-between the fibers, increasing the inter-fiber spacing, which is retained following sublimation. Structural characterization demonstrated cryo-scaffolds to have closer recapitulation of the trabecular meshwork, in terms of pore size, porosity, and thickness. The attachment of a healthy, human trabecular meshwork cell line (NTM5) to the scaffold was not influenced by the fabrication method. The main objective was to assess cell infiltration. Cryo-scaffolds supported cell penetration deep within their structure after seven days, whereas cells remained on the outer surface for conventional scaffolds. This study demonstrates the suitability of cryogenic electrospinning for the close recapitulation of trabecular meshwork and its potential as a 3D in vitro model and, in time, a tissue-engineered device.
Collapse
Affiliation(s)
- Devon J. Crouch
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, UK; (D.J.C.); (C.M.S.)
| | - Carl M. Sheridan
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, UK; (D.J.C.); (C.M.S.)
| | - Julia G. Behnsen
- Department of Mechanical, Materials, and Aerospace Engineering, University of Liverpool, Liverpool L69 6GB, UK;
| | - Raechelle A. D’Sa
- School of Engineering, University of Liverpool, Liverpool L69 3GH, UK;
| | - Lucy A. Bosworth
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, UK; (D.J.C.); (C.M.S.)
| |
Collapse
|
32
|
Puppi D, Braccini S, Battisti A, Manariti A, Pecorini G, Samal SK. Additive Manufacturing of Wet-Spun Polysulfone Medical Implants. ACS Biomater Sci Eng 2023; 9:5418-5429. [PMID: 37691546 DOI: 10.1021/acsbiomaterials.3c00711] [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: 09/12/2023]
Abstract
Research on additive manufacturing (AM) of high-performance polymers provides novel materials and technologies for advanced applications in different sectors, such as aerospace and biomedical engineering. The present article is contextualized in this research trend by describing a novel AM protocol for processing a polysulfone (PSU)/N-methyl-2-pyrrolidone (NMP) solution into medical implant prototypes. In particular, an AM technique involving the patterned deposition of the PSU/NMP mixture in a coagulation bath was employed to fabricate PSU implants with different predefined shape, fiber diameter, and macropore size. Scanning electron microscopy (SEM) analysis highlighted a fiber transversal cross-section morphology characterized by a dense external skin layer and an inner macroporous/microporous structure, as a consequence of the nonsolvent-induced polymer solidification process. Physical-chemical and thermal characterization of the fabricated samples demonstrated that PSU processing did not affect its macromolecular structure and glass-transition temperature, as well as that after post-processing PSU implants did not contain residual solvent or nonsolvent. Mechanical characterization showed that the developed PSU scaffold tensile and compressive modulus could be changed by varying the macroporous architecture. In addition, PSU scaffolds supported the in vitro adhesion and proliferation of the BALB/3T3 clone A31 mouse embryo cell line. These findings encourage further research on the suitability of the developed processing method for the fabrication of customized PSU implants.
Collapse
Affiliation(s)
- Dario Puppi
- Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Simona Braccini
- Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Antonella Battisti
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, p.zza San Silvestro 12, 56127 Pisa, Italy
| | - Antonella Manariti
- Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Gianni Pecorini
- Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Sangram Keshari Samal
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, Indian Council of Medical Research-Regional Medical Research Center, Bhubaneswar 751013, Odisha, India
| |
Collapse
|
33
|
Duan K, Mehwish N, Xu M, Zhu H, Hu J, Lin M, Yu L, Lee BH. Autoclavable Albumin-Based Cryogels with Uncompromising Properties. Gels 2023; 9:712. [PMID: 37754393 PMCID: PMC10530076 DOI: 10.3390/gels9090712] [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: 08/04/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/28/2023] Open
Abstract
The development of autoclavable hydrogels has been driven by the need for materials that can withstand the rigors of sterilization without compromising their properties or functionality. Many conventional hydrogels cannot withstand autoclave treatment owing to the breakdown of their composition or structure under the high-temperature and high-pressure environment of autoclaving. Here, the effect of autoclaving on the physical, mechanical, and biological properties of bovine serum albumin methacryloyl (BSAMA) cryogels at three protein concentrations (3, 5, and 10%) was extensively studied. We found that BSAMA cryogels at three concentrations remained little changed after autoclaving in terms of gross shape, pore structure, and protein secondary structure. Young's modulus of autoclaved BSAMA cryogels (BSAMAA) at low concentrations (3 and 5%) was similar to that of BSAMA cryogels, whereas 10% BSAMAA exhibited a higher Young's modulus value, compared with 10% BSAMA. Interestingly, BSAMAA cryogels prolonged degradation. Importantly, cell viability, drug release, and hemolytic behaviors were found to be similar among the pre- and post-autoclaved cryogels. Above all, autoclaving proved to be more effective in sterilizing BSAMA cryogels from bacteria contamination than UV and ethanol treatments. Thus, autoclavable BSAMA cryogels with uncompromising properties would be useful for biomedical applications.
Collapse
Affiliation(s)
- Kairui Duan
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325011, China;
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China; (M.X.); (H.Z.); (J.H.); (M.L.)
| | - Nabila Mehwish
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China; (M.X.); (H.Z.); (J.H.); (M.L.)
| | - Mengdie Xu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China; (M.X.); (H.Z.); (J.H.); (M.L.)
| | - Hu Zhu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China; (M.X.); (H.Z.); (J.H.); (M.L.)
| | - Jiajun Hu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China; (M.X.); (H.Z.); (J.H.); (M.L.)
| | - Mian Lin
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China; (M.X.); (H.Z.); (J.H.); (M.L.)
| | - Lu Yu
- Department of Optometry, Wenzhou Medical University, Wenzhou 325035, China;
| | - Bae Hoon Lee
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325011, China;
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China; (M.X.); (H.Z.); (J.H.); (M.L.)
- Department of Optometry, Wenzhou Medical University, Wenzhou 325035, China;
| |
Collapse
|
34
|
Yang Z, Wang B, Liu W, Li X, Liang K, Fan Z, Li JJ, Niu Y, He Z, Li H, Wang D, Lin J, Du Y, Lin J, Xing D. In situ self-assembled organoid for osteochondral tissue regeneration with dual functional units. Bioact Mater 2023; 27:200-215. [PMID: 37096194 PMCID: PMC10121637 DOI: 10.1016/j.bioactmat.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/01/2023] [Accepted: 04/02/2023] [Indexed: 04/26/2023] Open
Abstract
The regeneration of hierarchical osteochondral units is challenging due to difficulties in inducing spatial, directional and controllable differentiation of mesenchymal stem cells (MSCs) into cartilage and bone compartments. Emerging organoid technology offers new opportunities for osteochondral regeneration. In this study, we developed gelatin-based microcryogels customized using hyaluronic acid (HA) and hydroxyapatite (HYP), respectively for inducing cartilage and bone regeneration (denoted as CH-Microcryogels and OS-Microcryogels) through in vivo self-assembly into osteochondral organoids. The customized microcryogels showed good cytocompatibility and induced chondrogenic and osteogenic differentiation of MSCs, while also demonstrating the ability to self-assemble into osteochondral organoids with no delamination in the biphasic cartilage-bone structure. Analysis by mRNA-seq showed that CH-Microcryogels promoted chondrogenic differentiation and inhibited inflammation, while OS-Microcryogels facilitated osteogenic differentiation and suppressed the immune response, by regulating specific signaling pathways. Finally, the in vivo engraftment of pre-differentiated customized microcryogels into canine osteochondral defects resulted in the spontaneous assembly of an osteochondral unit, inducing simultaneous regeneration of both articular cartilage and subchondral bone. In conclusion, this novel approach for generating self-assembling osteochondral organoids utilizing tailor-made microcryogels presents a highly promising avenue for advancing the field of tissue engineering.
Collapse
Affiliation(s)
- Zhen Yang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Bin Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 10081, China
| | - Xiaoke Li
- Department of Orthopedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Zejun Fan
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, Australia
| | - Yudi Niu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Zihao He
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Hui Li
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Jianhao Lin
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Dan Xing
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| |
Collapse
|
35
|
Vidane AS, Nunes FC, Ferreira JA, Fukumasu H, Freitas SH, Pallone EMJA, Ambrósio CE. Biocompatibility and interaction of porous alumina-zirconia scaffolds with adipose-derived mesenchymal stem cells for bone tissue regeneration. Heliyon 2023; 9:e20128. [PMID: 37809419 PMCID: PMC10559935 DOI: 10.1016/j.heliyon.2023.e20128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Replacement of bone defects with bone graft or implant is an important therapeutic strategy that has been used in routine practice. However, the identification of biomaterials that can mimic natural bone properties and serve as bone substitutes remains a major challenge. In this context, alumina-zirconia (Al2O3/ZrO2) nanocomposites emerge as potential alternatives for biomedical applications, owing to their high mechanical strength, wear resistance, and biocompatibility. In this sense, in this study, we prepared porous Al2O3/ZrO2 nanocomposites (scaffolds) using the gelcasting method and biomimetically coated them with calcium phosphate (CaP). We evaluated the biocompatibility of the scaffolds using the quantitative MTT cytotoxicity test in L929 cells. Moreover, rabbit adipose-derived mesenchymal stem cells (rADMSCs) were seeded with CaP-containing and CaP-free porous samples to evaluate cell proliferation and cell-scaffold interaction in vitro. Our results showed that the Al2O3/ZrO2 scaffolds were non-cytotoxic, and there were no significant differences between CaP-containing and CaP-free scaffolds in terms of cell growth and adhesion. In contrast, when co-cultured with rADMSCs, the scaffolds enhanced cell proliferation and cell adhesion. The rADMSCs adhered and migrated through the pores of the scaffold and anchored to different poles with differentiated elongated structures. These results suggest osteogenic differentiation of rADMSCs in response to mechanical loading of Al2O3/ZrO2 scaffolds. Therefore, we conclude that Al2O3/ZrO2 scaffolds have demonstrated significant implications in bone tissue engineering and are valuable biomaterials for bone replacement.
Collapse
Affiliation(s)
- Atanasio S. Vidane
- Department of Clinics, Veterinary Faculty, Eduardo Mondlane University, Maputo, Mozambique
| | - Fabio C. Nunes
- Department of Biosystems Engineering, Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Julieta A. Ferreira
- Department of Biosystems Engineering, Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Heidge Fukumasu
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Silvio H. Freitas
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Eliria MJA. Pallone
- Department of Biosystems Engineering, Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Carlos E. Ambrósio
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Chen L, Zhou C, Jiang C, Huang X, Liu Z, Zhang H, Liang W, Zhao J. Translation of nanotechnology-based implants for orthopedic applications: current barriers and future perspective. Front Bioeng Biotechnol 2023; 11:1206806. [PMID: 37675405 PMCID: PMC10478008 DOI: 10.3389/fbioe.2023.1206806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/21/2023] [Indexed: 09/08/2023] Open
Abstract
The objective of bioimplant engineering is to develop biologically compatible materials for restoring, preserving, or altering damaged tissues and/or organ functions. The variety of substances used for orthopedic implant applications has been substantially influenced by modern material technology. Therefore, nanomaterials can mimic the surface properties of normal tissues, including surface chemistry, topography, energy, and wettability. Moreover, the new characteristics of nanomaterials promote their application in sustaining the progression of many tissues. The current review establishes a basis for nanotechnology-driven biomaterials by demonstrating the fundamental design problems that influence the success or failure of an orthopedic graft, cell adhesion, proliferation, antimicrobial/antibacterial activity, and differentiation. In this context, extensive research has been conducted on the nano-functionalization of biomaterial surfaces to enhance cell adhesion, differentiation, propagation, and implant population with potent antimicrobial activity. The possible nanomaterials applications (in terms of a functional nanocoating or a nanostructured surface) may resolve a variety of issues (such as bacterial adhesion and corrosion) associated with conventional metallic or non-metallic grafts, primarily for optimizing implant procedures. Future developments in orthopedic biomaterials, such as smart biomaterials, porous structures, and 3D implants, show promise for achieving the necessary characteristics and shape of a stimuli-responsive implant. Ultimately, the major barriers to the commercialization of nanotechnology-derived biomaterials are addressed to help overcome the limitations of current orthopedic biomaterials in terms of critical fundamental factors including cost of therapy, quality, pain relief, and implant life. Despite the recent success of nanotechnology, there are significant hurdles that must be overcome before nanomedicine may be applied to orthopedics. The objective of this review was to provide a thorough examination of recent advancements, their commercialization prospects, as well as the challenges and potential perspectives associated with them. This review aims to assist healthcare providers and researchers in extracting relevant data to develop translational research within the field. In addition, it will assist the readers in comprehending the scope and gaps of nanomedicine's applicability in the orthopedics field.
Collapse
Affiliation(s)
- Long Chen
- Department of Orthopedics, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, China
| | - Chanyi Jiang
- Department of Pharmacy, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Xiaogang Huang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| | - Zunyong Liu
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| | - Hengjian Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| |
Collapse
|
37
|
Vijayan V, Sreekumar S, Ahina KM, Lakra R, Kiran MS. Lanthanum Oxide Nanoparticles Reinforced Collagen ƙ-Carrageenan Hydroxyapatite Biocomposite as Angio-Osteogenic Biomaterial for In Vivo Osseointegration and Bone Repair. Adv Biol (Weinh) 2023; 7:e2300039. [PMID: 37080950 DOI: 10.1002/adbi.202300039] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/22/2023] [Indexed: 04/22/2023]
Abstract
A composite biomatrix fabricated with collagen, ƙ-carrageenan, hydroxyapatite reinforced with lanthanum oxide nanoparticles is explored as proangiogenic and osteogenic bone tissue repair biomaterial. The biomatrix shows increased physical and biological stability as observed from proteolytic degradation and thermal stability studies. The addition of lanthanum oxide nanoparticles facilitates good osseointegration coupled with simultaneous activation of proangiogenic properties to act as a bone mimicking material. The minimal level of reactive oxygen species and superior cytocompatibility help the as-synthesized biomatrix in achieving capillary migration into the bone micro environment. The composite biomatrix upregulates the expression of VEGF, VEGF-R2 genes in endothelial cells and osteopontin, osteocalcin in osteoblasts cells, respectively. The in vivo hard tissue repair experiment conducted in a rat model shows complete healing of the bone defect by eight weeks with the application of collagen-ƙ-carrageenan-hydroxyapatite-lanthanum oxide nanoparticle biomaterial when compared to the biomaterial made out of individual constituents alone. The biomaterial matrix gets biointegrated into the bone tissue and exerts its therapeutic value in bringing a faster osseo repair process. The study shows the feasibility of using rare-earth metal nanoparticles in combination with protein-polysaccharide biopolymers for bone regeneration.
Collapse
Affiliation(s)
- Vinu Vijayan
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
- University of Madras, Chennai, Tamil Nadu, 600005, India
| | - Sreelekshmi Sreekumar
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kannoth Madappurakkal Ahina
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
| | - Rachita Lakra
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
| | - Manikantan Syamala Kiran
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
- University of Madras, Chennai, Tamil Nadu, 600005, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
38
|
Liu Y, Guo Q, Zhang X, Wang Y, Mo X, Wu T. Progress in Electrospun Fibers for Manipulating Cell Behaviors. ADVANCED FIBER MATERIALS 2023; 5:1241-1272. [DOI: 10.1007/s42765-023-00281-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/08/2023] [Indexed: 01/06/2025]
|
39
|
Yazdanpanah Z, Sharma NK, Raquin A, Cooper DML, Chen X, Johnston JD. Printing tissue-engineered scaffolds made of polycaprolactone and nano-hydroxyapatite with mechanical properties appropriate for trabecular bone substitutes. Biomed Eng Online 2023; 22:73. [PMID: 37474951 PMCID: PMC10360269 DOI: 10.1186/s12938-023-01135-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Bone tissue engineering, based on three-dimensional (3D) printing technology, has emerged as a promising approach to treat bone defects using scaffolds. The objective of this study was to investigate the influence of porosity and internal structure on the mechanical properties of scaffolds. METHODS We fabricated composite scaffolds (which aimed to replicate trabecular bone) from polycaprolactone (PCL) reinforced with 30% (wt.) nano-hydroxyapatite (nHAp) by extrusion printing. Scaffolds with various porosities were designed and fabricated with and without an interlayer offset, termed as staggered and lattice structure, respectively. Mechanical compressive testing was performed to determine scaffold elastic modulus and yield strength. Linear regression was used to evaluate mechanical properties as a function of scaffold porosity. RESULTS Different relationships between mechanical properties and porosities were noted for the staggered and lattice structures. For elastic moduli, the two relationships intersected (porosity = 55%) such that the lattice structure exhibited higher moduli with porosity values greater than the intersection point; vice versa for the staggered structure. The lattice structure exhibited higher yield strength at all porosities. Mechanical testing results also indicated elastic moduli and yield strength properties comparable to trabecular bone (elastic moduli: 14-165 MPa; yield strength: 0.9-10 MPa). CONCLUSIONS Taken together, this study demonstrates that scaffolds printed from PCL/30% (wt.) nHAp with lattice and staggered structure offer promise for treating trabecular bone defects. This study identified the effect of porosity and internal structure on scaffold mechanical properties and provided suggestions for developing scaffolds with mechanical properties for substituting trabecular bone.
Collapse
Affiliation(s)
- Zahra Yazdanpanah
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada.
| | - Nitin Kumar Sharma
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Alice Raquin
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Institut Catholique Des Arts Et Métiers, 85000, La Roche-Sur-Yon, France
| | - David M L Cooper
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - James D Johnston
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
40
|
Li W, Wu Y, Zhang X, Wu T, Huang K, Wang B, Liao J. Self-healing hydrogels for bone defect repair. RSC Adv 2023; 13:16773-16788. [PMID: 37283866 PMCID: PMC10240173 DOI: 10.1039/d3ra01700a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/24/2023] [Indexed: 06/08/2023] Open
Abstract
Severe bone defects can be caused by various factors, such as tumor resection, severe trauma, and infection. However, bone regeneration capacity is limited up to a critical-size defect, and further intervention is required. Currently, the most common clinical method to repair bone defects is bone grafting, where autografts are the "gold standard." However, the disadvantages of autografts, including inflammation, secondary trauma and chronic disease, limit their application. Bone tissue engineering (BTE) is an attractive strategy for repairing bone defects and has been widely researched. In particular, hydrogels with a three-dimensional network can be used as scaffolds for BTE owing to their hydrophilicity, biocompatibility, and large porosity. Self-healing hydrogels respond rapidly, autonomously, and repeatedly to induced damage and can maintain their original properties (i.e., mechanical properties, fluidity, and biocompatibility) following self-healing. This review focuses on self-healing hydrogels and their applications in bone defect repair. Moreover, we discussed the recent progress in this research field. Despite the significant existing research achievements, there are still challenges that need to be addressed to promote clinical research of self-healing hydrogels in bone defect repair and increase the market penetration.
Collapse
Affiliation(s)
- Weiwei Li
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Yanting Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Xu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Tingkui Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu 610041 China
| | - Kangkang Huang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu 610041 China
| | - Beiyu Wang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu 610041 China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| |
Collapse
|
41
|
Rodimova S, Mozherov A, Elagin V, Karabut M, Shchechkin I, Kozlov D, Krylov D, Gavrina A, Kaplin V, Epifanov E, Minaev N, Bardakova K, Solovieva A, Timashev P, Zagaynova E, Kuznetsova D. FLIM imaging revealed spontaneous osteogenic differentiation of stem cells on gradient pore size tissue-engineered constructs. Stem Cell Res Ther 2023; 14:81. [PMID: 37046354 PMCID: PMC10091689 DOI: 10.1186/s13287-023-03307-6] [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: 09/06/2022] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND There is an urgent clinical need for targeted strategies aimed at the treatment of bone defects resulting from fractures, infections or tumors. 3D scaffolds represent an alternative to allogeneic MSC transplantation, due to their mimicry of the cell niche and the preservation of tissue structure. The actual structure of the scaffold itself can affect both effective cell adhesion and its osteoinductive properties. Currently, the effects of the structural heterogeneity of scaffolds on the behavior of cells and tissues at the site of damage have not been extensively studied. METHODS Both homogeneous and heterogeneous scaffolds were generated from poly(L-lactic acid) methacrylated in supercritical carbon dioxide medium and were fabricated by two-photon polymerization. The homogeneous scaffolds consist of three layers of cylinders of the same diameter, whereas the heterogeneous (gradient pore sizes) scaffolds contain the middle layer of cylinders of increased diameter, imitating the native structure of spongy bone. To evaluate the osteoinductive properties of both types of scaffold, we performed in vitro and in vivo experiments. Multiphoton microscopy with fluorescence lifetime imaging microscopy was used for determining the metabolic states of MSCs, as a sensitive marker of cell differentiation. The results obtained from this approach were verified using standard markers of osteogenic differentiation and based on data from morphological analysis. RESULTS The heterogeneous scaffolds showed improved osteoinductive properties, accelerated the metabolic rearrangements associated with osteogenic differentiation, and enhanced the efficiency of bone tissue recovery, thereby providing for both the development of appropriate morphology and mineralization. CONCLUSIONS The authors suggest that the heterogeneous tissue constructs are a promising tool for the restoration of bone defects. And, furthermore, that our results demonstrate that the use of label-free bioimaging methods can be considered as an effective approach for intravital assessment of the efficiency of differentiation of MSCs on scaffolds.
Collapse
Affiliation(s)
- Svetlana Rodimova
- N. I. Lobachevsky Nizhny Novgorod National Research State University, 23 Gagarina Ave., Nizhny Novgorod, Russia, 603022.
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000.
| | - Artem Mozherov
- N. I. Lobachevsky Nizhny Novgorod National Research State University, 23 Gagarina Ave., Nizhny Novgorod, Russia, 603022
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000
| | - Vadim Elagin
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000
| | - Maria Karabut
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000
| | - Ilya Shchechkin
- N. I. Lobachevsky Nizhny Novgorod National Research State University, 23 Gagarina Ave., Nizhny Novgorod, Russia, 603022
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000
| | - Dmitry Kozlov
- N. I. Lobachevsky Nizhny Novgorod National Research State University, 23 Gagarina Ave., Nizhny Novgorod, Russia, 603022
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000
| | - Dmitry Krylov
- N. I. Lobachevsky Nizhny Novgorod National Research State University, 23 Gagarina Ave., Nizhny Novgorod, Russia, 603022
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000
| | - Alena Gavrina
- N. I. Lobachevsky Nizhny Novgorod National Research State University, 23 Gagarina Ave., Nizhny Novgorod, Russia, 603022
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000
| | - Vladislav Kaplin
- Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, 4 Kosygina St, Moscow, Russia, 119991
| | - Evgenii Epifanov
- Research Center "Crystallography and Photonics", Institute of Photonic Technologies, Russian Academy of Sciences, 2 Pionerskaya St, Troitsk, Moscow, Russia, 108840
| | - Nikita Minaev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya Str, Moscow, Russia, 119991
| | - Ksenia Bardakova
- Research Center "Crystallography and Photonics", Institute of Photonic Technologies, Russian Academy of Sciences, 2 Pionerskaya St, Troitsk, Moscow, Russia, 108840
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya Str, Moscow, Russia, 119991
| | - Anna Solovieva
- Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, 4 Kosygina St, Moscow, Russia, 119991
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya Str, Moscow, Russia, 119991
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, 8-2 Trubetskaya Str, Moscow, Russia, 119991
| | - Elena Zagaynova
- N. I. Lobachevsky Nizhny Novgorod National Research State University, 23 Gagarina Ave., Nizhny Novgorod, Russia, 603022
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000
| | - Daria Kuznetsova
- N. I. Lobachevsky Nizhny Novgorod National Research State University, 23 Gagarina Ave., Nizhny Novgorod, Russia, 603022
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., Nizhny Novgorod, Russia, 603000
| |
Collapse
|
42
|
Cook KR, Head D, Dougan L. Modelling network formation in folded protein hydrogels by cluster aggregation kinetics. SOFT MATTER 2023; 19:2780-2791. [PMID: 36988480 DOI: 10.1039/d3sm00111c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Globular folded protein-based hydrogels are becoming increasingly attractive due to their specific biological functionality, as well as their responsiveness to stimuli. By modelling folded proteins as colloids, there are rich opportunities to explore network formation mechanisms in protein hydrogels that negate the need for computationally expensive simulations which capture the full complexity of proteins. Here we present a kinetic lattice-based model which simulates the formation of irreversibly chemically crosslinked, folded protein-based hydrogels. We identify the critical point of gel percolation, explore the range of network regimes covering diffusion-limited to reaction-limited cluster aggregation (DLCA and RLCA, respectively) network formation mechanisms and predict the final network structure, fractal dimensions and final gel porosity. We reveal a crossover between DLCA and RLCA mechanisms as a function of protein volume fraction and show how the final network structure is governed by the structure at the percolation point, regardless of the broad variation of non-percolating cluster masses observed across all systems. An analysis of the pore size distribution in the final network structures reveals that, approaching RLCA, gels have larger maximal pores than the DLCA counterparts for both volume fractions studied. This general kinetic model and the analysis tools generate predictions of network structure and concurrent porosity over a broad range of experimentally controllable parameters that are consistent with current expectations and understanding of experimental results.
Collapse
Affiliation(s)
- Kalila R Cook
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, UK.
| | - David Head
- School of Computing, University of Leeds, Leeds, UK
| | - Lorna Dougan
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, UK.
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| |
Collapse
|
43
|
A flexible design framework to design graded porous bone scaffolds with adjustable anisotropic properties. J Mech Behav Biomed Mater 2023; 140:105727. [PMID: 36801781 DOI: 10.1016/j.jmbbm.2023.105727] [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: 06/20/2022] [Revised: 01/10/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023]
Abstract
Since the success of bone regenerative medicine depends on scaffold morphological and mechanical properties, numerous scaffolds designs have been proposed in the last decade, including graded structures that are suited to enhance tissue ingrowth. Most of these structures are based either on foams with a random pore definition, or on the periodic repetition of a unit cell (UC). These approaches are limited by the range of target porosities and obtained effective mechanical properties, and do not permit to easily generate a pore size gradient from the core to the periphery of the scaffold. In opposition, the objective of the present contribution is to propose a flexible design framework to generate various three-dimensional (3D) scaffolds structures including cylindrical graded scaffolds from the definition of a UC by making use of a non-periodic mapping. Conformal mappings are firstly used to generate graded circular cross-sections, while 3D structures are then obtained by stacking the cross-sections with or without a twist between different scaffold layers. The effective mechanical properties of different scaffold configurations are presented and compared using an energy-based efficient numerical method, pointing out the versatility of the design procedure to separately govern longitudinal and transverse anisotropic scaffold properties. Among these configurations, a helical structure exhibiting couplings between transverse and longitudinal properties is proposed and permits to extend the adaptability of the proposed framework. In order to investigate the capacity of common additive manufacturing techniques to fabricate the proposed structures, a subset of these configurations is elaborated using a standard SLA setup, and subjected to experimental mechanical testing. Despite observed geometric differences between the initial design and the actual obtained structures, the effective properties are satisfyingly predicted by the proposed computational method. Promising perspectives are offered concerning the design of self-fitting scaffolds with on-demand properties depending on the clinical application.
Collapse
|
44
|
Geoghegan N, O'Loughlin M, Delaney C, Rochfort KD, Kennedy M, Kolagatla S, Podhorska L, Rodriguez BJ, Florea L, Kelleher SM. Controlled degradation of polycaprolactone-based micropillar arrays. Biomater Sci 2023; 11:3077-3091. [PMID: 36876330 PMCID: PMC10152922 DOI: 10.1039/d3bm00165b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Herein we demonstrate the fabrication of arrays of micropillars, achieved through the combination of direct laser writing and nanoimprint lithography. By combining two diacrylate monomers, polycaprolactone dimethacrylate (PCLDMA) and 1,6-hexanediol diacrylate (HDDA), two copolymer formulations that, owing to the varying ratios of the hydrolysable ester functionalities present in the polycaprolactone moiety, can be degraded in the presence of base in a controllable manner. As such, the degradation of the micropillars can be tuned over several days as a function of PCLDMA concentration within the copolymer formulations, and the topography greatly varied over a short space of time, as visualised using scanning electron microscopy and atomic force microscopy. Crosslinked neat HDDA was used as a control material, demonstrating that the presence of the PCL was responsible for the ability of the microstructures to degrade in the controlled manner. In addition, the mass loss of the crosslinked materials was minimal, demonstrating the degradation of microstructured surfaces without loss of bulk properties was possible. Moreover, the compatibility of these crosslinked materials with mammalian cells was explored. The influence of both indirect and direct contact of the materials with A549 cells was assessed by profiling indices reflective of cytotoxicity such as morphology, adhesion, metabolic activity, oxidative balance, and release of injury markers. No significant changes in the aforementioned profile were observed in the cells cultured under these conditions for up to 72 h, with the cell-material interaction suggesting these materials may have potential in microfabrication contexts towards biomedical application purposes.
Collapse
Affiliation(s)
- Niamh Geoghegan
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland. .,CURAM, Science Foundation Ireland Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Mark O'Loughlin
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Colm Delaney
- School of Chemistry and AMBER, the SFI Research Centre for Advanced Materials and BioEngineering Research, Trinity College Dublin, the University of Dublin, College Green, Dublin 2, Ireland
| | - Keith D Rochfort
- School of Nursing, Psychotherapy, and Community Health, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Meabh Kennedy
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Srikanth Kolagatla
- School of Chemistry and AMBER, the SFI Research Centre for Advanced Materials and BioEngineering Research, Trinity College Dublin, the University of Dublin, College Green, Dublin 2, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lucia Podhorska
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Brian J Rodriguez
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Larisa Florea
- School of Chemistry and AMBER, the SFI Research Centre for Advanced Materials and BioEngineering Research, Trinity College Dublin, the University of Dublin, College Green, Dublin 2, Ireland
| | - Susan M Kelleher
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland. .,CURAM, Science Foundation Ireland Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.,School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
45
|
Guo Y, Hu Z, Chen J, Zhang Z, Liu Q, Li J, Yang J, Ma Z, Zhao J, Hu J, Wu J, Chen Z. Injectable TG-linked recombinant human collagen hydrogel loaded with bFGF for rat cranial defect repair. Int J Biol Macromol 2023; 236:123864. [PMID: 36871688 DOI: 10.1016/j.ijbiomac.2023.123864] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
The basic fibroblast growth factor (bFGF) plays a significant role in promoting the process of bone repair, but bFGF cannot keep its biological activity stable under normal physiological conditions. Therefore, the development of better biomaterials to carry bFGF remains a challenge for bone repair and regeneration. Here we designed a novel recombinant human collagen (rhCol), which could be cross-linked by transglutaminase (TG) and loaded bFGF to prepare rhCol/bFGF hydrogels. The rhCol hydrogel possessed a porous structure and good mechanical properties. The assays, including cell proliferation, migration, and adhesion assay, were performed to evaluate the biocompatibility of rhCol/bFGF and the results demonstrated that the rhCol/bFGF promoted cell proliferation, migration and adhesion. The rhCol/bFGF hydrogel degraded and released bFGF controllably, enhancing utilization rate of bFGF and allowing osteoinductive activity. The results of RT-qPCR and immunofluorescence staining also proved that rhCol/bFGF promoted expression of bone-related proteins. The rhCol/bFGF hydrogels were applied in the cranial defect in rats and the results confirmed that it accelerates bone defect repair. In conclusion, rhCol/bFGF hydrogel has excellent biomechanical properties and can continuously release bFGF to promote bone regeneration, suggesting that rhCol/bFGF hydrogel is a potential scaffold in clinic application.
Collapse
Affiliation(s)
- Yayuan Guo
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Zeyu Hu
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Jilong Chen
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Zhen Zhang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Qian Liu
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Juan Li
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Jiaojiao Yang
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Zihan Ma
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Jing Zhao
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Jingyan Hu
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Jiawei Wu
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Zhuoyue Chen
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China.
| |
Collapse
|
46
|
Tardelli JDC, de Barros Ciribelli Alves BM, da Costa Valente ML, dos Reis AC. Influence of the modulus of elasticity of dental implants on the distribution of stresses in the alveolar bone by the finite element method: a systematic review. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY, MEDICINE, AND PATHOLOGY 2023. [DOI: 10.1016/j.ajoms.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
47
|
Capêto AP, Azevedo-Silva J, Sousa S, Pintado M, Guimarães AS, Oliveira ALS. Synthesis of Bio-Based Polyester from Microbial Lipidic Residue Intended for Biomedical Application. Int J Mol Sci 2023; 24:4419. [PMID: 36901850 PMCID: PMC10003017 DOI: 10.3390/ijms24054419] [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: 01/04/2023] [Revised: 02/13/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
In the last decade, selectively tuned bio-based polyesters have been increasingly used for their clinical potential in several biomedical applications, such as tissue engineering, wound healing, and drug delivery. With a biomedical application in mind, a flexible polyester was produced by melt polycondensation using the microbial oil residue collected after the distillation of β-farnesene (FDR) produced industrially by genetically modified yeast, Saccharomyces cerevisiae. After characterization, the polyester exhibited elongation up to 150% and presented Tg of -51.2 °C and Tm of 169.8 °C. In vitro degradation revealed a mass loss of about 87% after storage in PBS solution for 11 weeks under accelerated conditions (40 °C, RH = 75%). The water contact angle revealed a hydrophilic character, and biocompatibility with skin cells was demonstrated. 3D and 2D scaffolds were produced by salt-leaching, and a controlled release study at 30 °C was performed with Rhodamine B base (RBB, 3D) and curcumin (CRC, 2D), showing a diffusion-controlled mechanism with about 29.3% of RBB released after 48 h and 50.4% of CRC after 7 h. This polymer offers a sustainable and eco-friendly alternative for the potential use of the controlled release of active principles for wound dressing applications.
Collapse
Affiliation(s)
- Ana P. Capêto
- Centro de Biotecnologia e Química Fina (CBQF)-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo de Botelho 1327, 4169-005 Porto, Portugal
| | - João Azevedo-Silva
- Centro de Biotecnologia e Química Fina (CBQF)-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo de Botelho 1327, 4169-005 Porto, Portugal
| | - Sérgio Sousa
- Centro de Biotecnologia e Química Fina (CBQF)-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo de Botelho 1327, 4169-005 Porto, Portugal
| | - Manuela Pintado
- Centro de Biotecnologia e Química Fina (CBQF)-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo de Botelho 1327, 4169-005 Porto, Portugal
| | - Ana S. Guimarães
- CONSTRUCT, Faculdade de Engenharia do Porto (FEUP), Universidade do Porto, Rua Doutor Roberto Frias, 4200-465 Porto, Portugal
| | - Ana L. S. Oliveira
- Centro de Biotecnologia e Química Fina (CBQF)-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo de Botelho 1327, 4169-005 Porto, Portugal
| |
Collapse
|
48
|
Graphene-Based Materials in Dental Applications: Antibacterial, Biocompatible, and Bone Regenerative Properties. Int J Biomater 2023; 2023:8803283. [PMID: 36819211 PMCID: PMC9929215 DOI: 10.1155/2023/8803283] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 02/10/2023] Open
Abstract
Graphene-based materials have been shown to have advantageous properties in biomedical and dental applications due to their high mechanical, physiochemical, antibacterial, and stem cell differentiating properties. Although graphene-based materials have displayed appropriate biocompatible properties when used in implant materials for orthopedic applications, little research has been performed to specifically test the biocompatibility of graphene for dental applications. The oral environment, compared to the body, varies greatly and must be considered when evaluating biocompatibility requirements for dental applications. This review will discuss in vitro and in vivo studies that assess graphene's cytotoxicity, antibacterial properties, and cell differentiation ability to evaluate the overall biocompatibility of graphene-based materials for dental applications. Particle shape, size, and concentration were found to be major factors that affected overall biocompatibility of graphene.
Collapse
|
49
|
Wu T, Wu Y, Cao Z, Zhao L, Lv J, Li J, Xu Y, Zhang P, Liu X, Sun Y, Cheng M, Tang K, Jiang X, Ling C, Yao Q, Zhu Y. Cell-free and cytokine-free self-assembling peptide hydrogel-polycaprolactone composite scaffolds for segmental bone defects. Biomater Sci 2023; 11:840-853. [PMID: 36512317 DOI: 10.1039/d2bm01609e] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Segmental bone defects over the self-healing threshold are a major challenge for orthopedics. Despite the advancements in clinical practice, traditional tissue engineering methods are limited by the addition of heterogeneous cells and cytokines, leading to carcinoma or other adverse effects. Here, we present a cell-free and cytokine-free strategy using an ECM-mimetic self-assembling peptide hydrogel (SAPH)- polycaprolactone (PCL) composite scaffold. The hydrophilic SAPH endows the rigid PCL scaffold with excellent biocompatibility and preference for osteogenesis induction. The autologous cells around the bone defect site immediately grew, proliferated, and secreted ECM and cytokines after contacting the implanted SAPH-PCL composite scaffold, and the bone repair of rabbit ulnar segmental bone defect was achieved in just six months. Quantitative proteomic analysis reveals that the SAPH-PCL composite scaffold accelerates osteoblastogenesis, osteoclastogenesis, and angiogenesis with moderate immune responses and negligible effects on pathological fibrosis. These findings have important implications for the potential clinical applications of the SAPH-PCL composite scaffold in patients with segmental bone defects and identify the mechanisms of action for accelerated segmental bone defect repair.
Collapse
Affiliation(s)
- Tong Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Yilun Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Lulu Zhao
- College of Pharmaceutical Sciences, Nanjing Tech University, 211816, Nanjing, China
| | - Jiayi Lv
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Jiayi Li
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Yue Xu
- College of Pharmaceutical Sciences, Nanjing Tech University, 211816, Nanjing, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Xu Liu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Min Cheng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Kexin Tang
- College of Pharmaceutical Sciences, Nanjing Tech University, 211816, Nanjing, China
| | - Xiao Jiang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Chen Ling
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Yishen Zhu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| |
Collapse
|
50
|
Liu YX, Chaparro FJ, Tian Z, Jia Y, Gosser J, Gaumer J, Ross L, Tafreshi H, Lannutti JJ. Visualization of porosity and pore size gradients in electrospun scaffolds using laser metrology. PLoS One 2023; 18:e0282903. [PMID: 36893193 PMCID: PMC9997878 DOI: 10.1371/journal.pone.0282903] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/24/2023] [Indexed: 03/10/2023] Open
Abstract
We applied a recently developed method, laser metrology, to characterize the influence of collector rotation on porosity gradients of electrospun polycaprolactone (PCL) widely investigated for use in tissue engineering. The prior- and post-sintering dimensions of PCL scaffolds were compared to derive quantitative, spatially-resolved porosity 'maps' from net shrinkage. Deposited on a rotating mandrel (200 RPM), the central region of deposition reaches the highest porosity, ~92%, surrounded by approximately symmetrical decreases to ~89% at the edges. At 1100 RPM, a uniform porosity of ~88-89% is observed. At 2000 RPM, the lowest porosity, ~87%, is found in the middle of the deposition, rebounding to ~89% at the edges. Using a statistical model of random fiber network, we demonstrated that these relatively small changes in porosity values produce disproportionately large variations in pore size. The model predicts an exponential dependence of pore size on porosity when the scaffold is highly porous (e.g., >80%) and, accordingly, the observed porosity variation is associated with dramatic changes in pore size and ability to accommodate cell infiltration. Within the thickest regions most likely to 'bottleneck' cell infiltration, pore size decreases from ~37 to 23 μm (38%) when rotational speeds increased from 200 to 2000 RPM. This trend is corroborated by electron microscopy. While faster rotational speeds ultimately overcome axial alignment induced by cylindrical electric fields associated with the collector geometry, it does so at the cost of eliminating larger pores favoring cell infiltration. This puts the bio-mechanical advantages associated with collector rotation-induced alignment at odds with biological goals. A more significant decrease in pore size from ~54 to ~19 μm (65%), well below the minimum associated with cellular infiltration, is observed from enhanced collector biases. Finally, similar predictions show that sacrificial fiber approaches are inefficient in achieving cell-permissive pore sizes.
Collapse
Affiliation(s)
- Yi-xiao Liu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
- * E-mail:
| | | | - Ziting Tian
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Yizhen Jia
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
| | - John Gosser
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Jeremy Gaumer
- Tosoh SMD, Inc., Grove City, OH, United States of America
| | - Liam Ross
- Columbus Academy, Gahanna, OH, United States of America
| | - Hooman Tafreshi
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC, United States of America
| | - John J. Lannutti
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States of America
- Center for Chronic Brain Injury Program, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|