1
|
Varshney S, Dwivedi A, Pandey V. Bioprinting techniques for regeneration of oral and craniofacial tissues: Current advances and future prospects. J Oral Biol Craniofac Res 2025; 15:331-346. [PMID: 40027866 PMCID: PMC11870160 DOI: 10.1016/j.jobcr.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/12/2024] [Accepted: 01/18/2025] [Indexed: 03/05/2025] Open
Abstract
Background Regenerative dentistry aims to reinstate, fix, renew, and regrow tissues within the oral and craniofacial domain. Existing regenerative methods are based on insights into tissue biology or disease processes that lead to tissue degradation. However, achieving complete and functional Tissue regeneration remains a primary challenge in real-world medical scenarios. Aim The review focuses on the application of bioprinting techniques for rejuvenating intricate Oral and craniofacial tissues, such as craniofacial bone, periodontal ligament, cementum, dental pulp, temporomandibular joint cartilage, and whole teeth. Methods Bioprinting, a cutting-edge technology in regenerative dentistry, strives to create entirely new Functional tissues and organs. This approach merges principles from engineering and biology to produce three-dimensional biologically operational constructs containing bioactive substances, Living cells and cell clusters using automated bioprinters. The review summarizes the outcomes achieved through bioprinting techniques in both in vitro (laboratory experiments) and in vivo (Studies on living organisms) experiments. Result The emergence of this innovative tissue engineering technology has yielded highly promising outcomes during the experimental stages. Conclusion These promising experimental results necessitate replication through human clinical trials to ascertain the viability of bioprinting techniques for mainstream clinical implementation in regenerative dentistry.
Collapse
Affiliation(s)
- Shailesh Varshney
- Department of Periodontology, School of Dental Sciences, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Anshuman Dwivedi
- ,Department of Stem Cells & Regenerative Medicine, Santosh, University, Ghaziabad, Uttar Pradesh, India
| | - Vibha Pandey
- ,Department of Psychology, Himalayan, Garhwal University, Uttarakhand, India
| |
Collapse
|
2
|
Gezek M, Altunbek M, Torres Gouveia ME, Camci-Unal G. 3D Printed Eggshell Microparticle-Laden Thermoplastic Scaffolds for Bone Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2024; 16:32957-32970. [PMID: 38885611 DOI: 10.1021/acsami.4c02800] [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: 06/20/2024]
Abstract
Three-dimensional (3D) printing, an additive manufacturing technique, is increasingly used in the field of tissue engineering. The ability to create complex structures with high precision makes the 3D printing of this material a preferred method for constructing personalized and functional materials. However, the challenge lies in developing affordable and accessible materials with the desired physiochemical and biological properties. In this study, we used eggshell microparticles (ESPs), an example of bioceramic and unconventional biomaterials, to reinforce thermoplastic poly(ε-caprolactone) (PCL) scaffolds via extrusion-based 3D printing. The goal was to conceive a sustainable, affordable, and unique personalized medicine approach. The scaffolds were fabricated with varying concentrations of eggshells, ranging from 0 to 50% (w/w) in the PCL scaffolds. To assess the physicochemical properties, we employed scanning electron microscopy, Fourier-transform infrared spectroscopy, thermogravimetric analysis, differential scanning calorimetry, and X-ray diffraction analysis. Mechanical properties were evaluated through compression testing, and degradation kinetics were studied through accelerated degradation with the remaining mass ranging between 89.4 and 28.3%. In vitro, we evaluated the characteristics of the scaffolds using the MC3T3-E1 preosteoblasts over a 14 day period. In vitro characterization involved the use of the Alamar blue assay, confocal imaging, and real-time quantitative polymerase chain reaction. The results of this study demonstrate the potential of 3D printed biocomposite scaffolds, consisting of thermoplastic PCL reinforced with ESPs, as a promising alternative for bone-graft applications.
Collapse
Affiliation(s)
- Mert Gezek
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Mine Altunbek
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | | | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
3
|
Daskalakis E, Huang B, Hassan MH, Omar AM, Vyas C, Acar AA, Fallah A, Cooper G, Weightman A, Blunn G, Koç B, Bartolo P. In Vitro Evaluation of Pore Size Graded Bone Scaffolds with Different Material Composition. 3D PRINTING AND ADDITIVE MANUFACTURING 2024; 11:e718-e730. [PMID: 38689909 PMCID: PMC11057695 DOI: 10.1089/3dp.2022.0138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The demand for biomimetic and biocompatible scaffolds in equivalence of structure and material composition for the regeneration of bone tissue is relevantly high. This article is investigating a novel three-dimensional (3D) printed porous structure called bone bricks with a gradient pore size mimicking the structure of the bone tissue. Poly-ɛ-caprolactone (PCL) combined with ceramics such as hydroxyapatite (HA), β-tricalcium phosphate (TCP), and bioglass 45S5 were successfully mixed using a melt blending method and fabricated with the use of screw-assisted extrusion-based additive manufacturing system. Bone bricks containing the same material concentration (20 wt%) were biologically characterized through proliferation and differentiation tests. Scanning electron microscopy (SEM) was used to investigate the morphology of cells on the surface of bone bricks, whereas energy dispersive X-ray (EDX) spectroscopy was used to investigate the element composition on the surface of the bone bricks. Confocal imaging was used to investigate the number of differentiated cells on the surface of bone bricks. Proliferation results showed that bone bricks containing PCL/HA content are presenting higher proliferation properties, whereas differentiation results showed that bone bricks containing PCL/Bioglass 45S5 are presenting higher differentiation properties. Confocal imaging results showed that bone bricks containing PCL/Bioglass 45S5 are presenting a higher number of differentiated cells on their surface compared with the other material contents.
Collapse
Affiliation(s)
- Evangelos Daskalakis
- Department of Mechanical, Aerospace and Civil Engineering, The University of Manchester, Manchester, United Kingdom
| | - Boyang Huang
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| | - Mohamed H. Hassan
- Department of Mechanical, Aerospace and Civil Engineering, The University of Manchester, Manchester, United Kingdom
| | - Abdalla M. Omar
- Department of Mechanical, Aerospace and Civil Engineering, The University of Manchester, Manchester, United Kingdom
| | - Cian Vyas
- Department of Mechanical, Aerospace and Civil Engineering, The University of Manchester, Manchester, United Kingdom
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| | - Anil A. Acar
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Istanbul, Turkey
- SUNUM Nanotechnology Research Center, Sabanci University, Istanbul, Turkey
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Ali Fallah
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Istanbul, Turkey
- SUNUM Nanotechnology Research Center, Sabanci University, Istanbul, Turkey
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Glen Cooper
- Department of Mechanical, Aerospace and Civil Engineering, The University of Manchester, Manchester, United Kingdom
| | - Andrew Weightman
- Department of Mechanical, Aerospace and Civil Engineering, The University of Manchester, Manchester, United Kingdom
| | - Gordon Blunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Bahattin Koç
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Istanbul, Turkey
- SUNUM Nanotechnology Research Center, Sabanci University, Istanbul, Turkey
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Paulo Bartolo
- Department of Mechanical, Aerospace and Civil Engineering, The University of Manchester, Manchester, United Kingdom
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
4
|
Lin H, Zhang L, Zhang Q, Wang Q, Wang X, Yan G. Mechanism and application of 3D-printed degradable bioceramic scaffolds for bone repair. Biomater Sci 2023; 11:7034-7050. [PMID: 37782081 DOI: 10.1039/d3bm01214j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Bioceramics have attracted considerable attention in the field of bone repair because of their excellent osteogenic properties, degradability, and biocompatibility. To resolve issues regarding limited formability, recent studies have introduced 3D printing technology for the fabrication of bioceramic bone repair scaffolds. Nevertheless, the mechanisms by which bioceramics promote bone repair and clinical applications of 3D-printed bioceramic scaffolds remain elusive. This review provides an account of the fabrication methods of 3D-printed degradable bioceramic scaffolds. In addition, the types and characteristics of degradable bioceramics used in clinical and preclinical applications are summarized. We have also highlighted the osteogenic molecular mechanisms in biomaterials with the aim of providing a basis and support for future research on the clinical applications of degradable bioceramic scaffolds. Finally, new developments and potential applications of 3D-printed degradable bioceramic scaffolds are discussed with reference to experimental and theoretical studies.
Collapse
Affiliation(s)
- Hui Lin
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Liyun Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Qiyue Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Xue Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
| | - Guangqi Yan
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
| |
Collapse
|
5
|
Shiroud Heidari B, Lopez EM, Chen P, Ruan R, Vahabli E, Davachi SM, Granero-Moltó F, De-Juan-Pardo EM, Zheng M, Doyle B. Silane-modified hydroxyapatite nanoparticles incorporated into polydioxanone/poly(lactide- co-caprolactone) creates a novel toughened nanocomposite with improved material properties and in vivo inflammatory responses. Mater Today Bio 2023; 22:100778. [PMID: 37664796 PMCID: PMC10474235 DOI: 10.1016/j.mtbio.2023.100778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/13/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023] Open
Abstract
The interface tissue between bone and soft tissues, such as tendon and ligament (TL), is highly prone to injury. Although different biomaterials have been developed for TL regeneration, few address the challenges of the TL-bone interface. Here, we aim to develop novel hybrid nanocomposites based on poly(p-dioxanone) (PDO), poly(lactide-co-caprolactone) (LCL), and hydroxyapatite (HA) nanoparticles suitable for TL-bone interface repair. Nanocomposites, containing 3-10% of both unmodified and chemically modified hydroxyapatite (mHA) with a silane coupling agent. We then explored biocompatibility through in vitro and in vivo studies using a subcutaneous mouse model. Through different characterisation tests, we found that mHA increases tensile properties, creates rougher surfaces, and reduces crystallinity and hydrophilicity. Morphological observations indicate that mHA nanoparticles are attracted by PDO rather than LCL phase, resulting in a higher degradation rate for mHA group. We found that adding the 5% of nanoparticles gives a balance between the properties. In vitro experiments show that osteoblasts' activities are more affected by increasing the nanoparticle content compared with fibroblasts. Animal studies indicate that both HA and mHA nanoparticles (10%) can reduce the expression of pro-inflammatory cytokines after six weeks of implantation. In summary, this work highlights the potential of PDO/LCL/HA nanocomposites as an excellent biomaterial for TL-bone interface tissue engineering applications.
Collapse
Affiliation(s)
- Behzad Shiroud Heidari
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia
- School of Engineering, The University of Western Australia, Perth, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Emma Muinos Lopez
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Peilin Chen
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia
- School of Medicine, Monash University, VIC, Melbourne, Australia
| | - Rui Ruan
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia
| | - Ebrahim Vahabli
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia
- School of Engineering, The University of Western Australia, Perth, Australia
| | - Seyed Mohammad Davachi
- Department of Biology and Chemistry, Texas A&M International University, Laredo, TX, USA
| | - Froilán Granero-Moltó
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Elena M. De-Juan-Pardo
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia
- School of Engineering, The University of Western Australia, Perth, Australia
| | - Minghao Zheng
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Barry Doyle
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia
- School of Engineering, The University of Western Australia, Perth, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
6
|
Ahmadipour S, Varshosaz J, Hashemibeni B, Manshaei M, Safaeian L. In vivo assessment of bone repair by an injectable nanocomposite scaffold for local co-delivery of autologous platelet-rich plasma and calcitonin in rat model. Drug Dev Ind Pharm 2022; 48:98-108. [PMID: 35659167 DOI: 10.1080/03639045.2022.2087080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Background: Gellan gum is obtained from the bacterium Sphingomonas elodea and is a polysaccharide with carboxylic acid functional groups. The goal of this project was to investigate the osteoinductive effect of local administration of calcitonin through an injectable scaffold of gellan gum containing salmon calcitonin loaded in silsesquioxane nanoparticles, hydroxyapatite, and platelets rich plasma.Methods: The femur of rats was defected by creating a 2 × 5 mm2 hole using an electric drill. The defect was filled with an injectable hydrogel scaffold composed of gellan gum enriched with salmon calcitonin loaded in silsesquioxane nanoparticles, hydroxyapatite, platelets rich plasma and then the radiologic images were taken. Bone densitometry and the histologic studies were carried out by Hematoxylin & Eosin test. Biochemical analysis was done to measure the serum alkaline phosphatase (ALP), calcium, calcitonin concentration.Results: Healing of the bone defects and bone densitometry in the treated group by calcitonin-loaded scaffold was significantly higher (p < 0.05) and bone formation occupied 75% of the defect that was greater than other groups. Serum ALP and calcium levels in the scaffold-loaded calcitonin group were more than the other groups (p < 0.05). The osteogenic marker genes also increased significantly (p < 0.05) with free calcitonin and the scaffold.Conclusions: Gellan gum-based scaffold loaded with calcitonin may be considered a promising local treatment to progress bone formation in repairing of skeletal injuries.
Collapse
Affiliation(s)
- Saeedeh Ahmadipour
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Pharmaceutics, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences, Faculty of Medicine; Torabinejad Dental Research Center, Dental School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maziar Manshaei
- Dental research center, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Safaeian
- Department of Pharmacology and Toxicology, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
7
|
Bioactivity of star-shaped polycaprolactone/chitosan composite hydrogels for biomaterials. Int J Biol Macromol 2022; 212:420-431. [PMID: 35623458 DOI: 10.1016/j.ijbiomac.2022.05.139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 11/21/2022]
Abstract
Recently, our group reported the synthesis and fabrication of composite hydrogels of chitosan (CS) and star-shaped polycaprolactone (stPCL). The co-crosslink of modified stPCL with carboxyl at the end chain (stPCL-COOH) provided good mechanical properties and stability to the composite hydrogels. This research presents the bioactivities of composite hydrogels showing a potential candidate to develop biomaterials such as wound dressing and bone tissue engineering. The bioactivities were the antibacterial activity, cell viability, skin irritation, decomposability, and ability to attach ions for apatite nucleation. The results showed that all the composite hydrogels were completely decomposed within 2 days. The composite hydrogels had better antibacterial activity and higher efficiency to Gram-negative (Escherichia coli) than to Gram-positive (Staphylococcus epidermidis) bacteria. The composite hydrogels were studied for cell viability based on MTT assay and skin irritation on rabbit skin. The results indicated high cell survival more than 80% and no skin irritation. In addition, the results showed that calcium and phosphorous were preferentially attached to the composite hydrogel surface to grow apatite crystal (Ca/P ratio 1.86) compared to attaching to the chitosan hydrogel (Ca/P ratio 1.48) in 21 days of testing.
Collapse
|
8
|
Sivakumar PM, Yetisgin AA, Sahin SB, Demir E, Cetinel S. Bone tissue engineering: Anionic polysaccharides as promising scaffolds. Carbohydr Polym 2022; 283:119142. [DOI: 10.1016/j.carbpol.2022.119142] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 12/21/2022]
|
9
|
Abazari MF, Torabinejad S, Zare Karizi S, Enderami SE, Samadian H, Hajati-Birgani N, Norouzi S, Nejati F, Al bahash A, Mansouri V. Promoted osteogenic differentiation of human induced pluripotent stem cells using composited polycaprolactone/polyvinyl alcohol/carbopol nanofibrous scaffold. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
10
|
Joseph J, Parameswaran R, Gopalakrishna Panicker U. Recent advancements in blended and reinforced polymeric systems as bioscaffolds. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2066666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Jasmin Joseph
- Department of Chemistry, National Institute of Technology, Calicut, India
- Division of Polymeric Medical Devices, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Ramesh Parameswaran
- Division of Polymeric Medical Devices, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | | |
Collapse
|
11
|
Wang H, Hu B, Li H, Feng G, Pan S, Chen Z, Li B, Song J. Biomimetic Mineralized Hydroxyapatite Nanofiber-Incorporated Methacrylated Gelatin Hydrogel with Improved Mechanical and Osteoinductive Performances for Bone Regeneration. Int J Nanomedicine 2022; 17:1511-1529. [PMID: 35388269 PMCID: PMC8978691 DOI: 10.2147/ijn.s354127] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/15/2022] [Indexed: 12/16/2022] Open
Abstract
Purpose Methacrylic anhydride-modified gelatin (GelMA) hydrogels exhibit many beneficial biological features and are widely studied for bone tissue regeneration. However, deficiencies in the mechanical strength, osteogenic factors and mineral ions limit their application in bone defect regeneration. Incorporation of inorganic fillers into GelMA to improve its mechanical properties and bone regenerative ability has been one of the research hotspots. Methods In this work, hydroxyapatite nanofibers (HANFs) were prepared and mineralized in a simulated body fluid to make their components and structure more similar to those of natural bone apatite, and then different amounts of mineralized HANFs (m-HANFs) were incorporated into the GelMA hydrogel to form m-HANFs/GelMA composite hydrogels. The physicochemical properties, biocompatibility and bone regenerative ability of m-HANFs/GelMA were determined in vitro and in vivo. Results The results indicated that m-HANFs with high aspect ratio presented rough and porous surfaces coated with bone-like apatite crystals. The incorporation of biomimetic m-HANFs improved the biocompatibility, mechanical, swelling, degradation and bone regenerative performances of GelMA. However, the improvement in the performance of the composite hydrogel did not continuously increase as the amount of added m-HANFs increased, and the 15m-HANFs/GelMA group exhibited the best swelling and degradation performances and the best bone repair effect in vivo among all the groups. Conclusion The biomimetic m-HANFs/GelMA composite hydrogel can provide a novel option for bone tissue engineering in the future; however, it needs further investigations to optimize the proportions of m-HANFs and GelMA for improving the bone repair effect.
Collapse
Affiliation(s)
- He Wang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Bo Hu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Hong Li
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing, People’s Republic of China
| | - Ge Feng
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Shengyuan Pan
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Ziqi Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Bo Li
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing, People’s Republic of China
- Correspondence: Bo Li, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, People’s Republic of China, Tel +86-23-8886-0026, Fax +86-23-8886-0222, Email
| | - Jinlin Song
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
- Jinlin Song, College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, People’s Republic of China, Tel +86-23-8886-0026, Fax +86-23-8886-0222, Email
| |
Collapse
|
12
|
Murugan S, Parcha SR. Fabrication techniques involved in developing the composite scaffolds PCL/HA nanoparticles for bone tissue engineering applications. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:93. [PMID: 34379204 PMCID: PMC8357662 DOI: 10.1007/s10856-021-06564-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 05/28/2021] [Indexed: 06/04/2023]
Abstract
A fine-tuned combination of scaffolds, biomolecules, and mesenchymal stem cells (MSCs) is used in tissue engineering to restore the function of injured bone tissue and overcome the complications associated with its regeneration. For two decades, biomaterials have attracted much interest in mimicking the native extracellular matrix of bone tissue. To this aim, several approaches based on biomaterials combined with MSCs have been amply investigated. Recently, hydroxyapatite (HA) nanoparticles have been incorporated with polycaprolactone (PCL) matrix as a suitable substitute for bone tissue engineering applications. This review article aims at providing a brief overview on PCL/HA composite scaffold fabrication techniques such as sol-gel, rapid prototyping, electro-spinning, particulate leaching, thermally induced phase separation, and freeze-drying, as suitable approaches for tailoring morphological, mechanical, and biodegradability properties of the scaffolds for bone tissues. Among these methods, the 3D plotting method shows improvements in pore architecture (pore size of ≥600 µm and porosity of 92%), mechanical properties (higher than 18.38 MPa), biodegradability, and good bioactivity in bone tissue regeneration.
Collapse
Affiliation(s)
- Sivasankar Murugan
- Stem Cell Research Laboratory, Department of Biotechnology, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Sreenivasa Rao Parcha
- Stem Cell Research Laboratory, Department of Biotechnology, National Institute of Technology, Warangal, Telangana, 506004, India.
| |
Collapse
|
13
|
Moncal KK, Gudapati H, Godzik KP, Heo DN, Kang Y, Rizk E, Ravnic DJ, Wee H, Pepley DF, Ozbolat V, Lewis GS, Moore JZ, Driskell RR, Samson TD, Ozbolat IT. Intra-Operative Bioprinting of Hard, Soft, and Hard/Soft Composite Tissues for Craniomaxillofacial Reconstruction. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2010858. [PMID: 34421475 PMCID: PMC8376234 DOI: 10.1002/adfm.202010858] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Indexed: 05/20/2023]
Abstract
Reconstruction of complex craniomaxillofacial (CMF) defects is challenging due to the highly organized layering of multiple tissue types. Such compartmentalization necessitates the precise and effective use of cells and other biologics to recapitulate the native tissue anatomy. In this study, intra-operative bioprinting (IOB) of different CMF tissues, including bone, skin, and composite (hard/soft) tissues, is demonstrated directly on rats in a surgical setting. A novel extrudable osteogenic hard tissue ink is introduced, which induced substantial bone regeneration, with ≈80% bone coverage area of calvarial defects in 6 weeks. Using droplet-based bioprinting, the soft tissue ink accelerated the reconstruction of full-thickness skin defects and facilitated up to 60% wound closure in 6 days. Most importantly, the use of a hybrid IOB approach is unveiled to reconstitute hard/soft composite tissues in a stratified arrangement with controlled spatial bioink deposition conforming the shape of a new composite defect model, which resulted in ≈80% skin wound closure in 10 days and 50% bone coverage area at Week 6. The presented approach will be absolutely unique in the clinical realm of CMF defects and will have a significant impact on translating bioprinting technologies into the clinic in the future.
Collapse
Affiliation(s)
- Kazim K Moncal
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Hemanth Gudapati
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kevin P Godzik
- Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Dong N Heo
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Youngnam Kang
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Elias Rizk
- Department of Neurosurgery, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Dino J Ravnic
- Department of Surgery, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Hwabok Wee
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - David F Pepley
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Veli Ozbolat
- Mechanical Engineering Department, Ceyhan Engineering Faculty, Cukurova University, Adana 01950, Turkey
| | - Gregory S Lewis
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jason Z Moore
- Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Ryan R Driskell
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | - Thomas D Samson
- Department of Neurosurgery, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
14
|
Abdollahi Boraei SB, Nourmohammadi J, Bakhshandeh B, Dehghan MM, Gholami H, Gonzalez Z, Sanchez-Herencia AJ, Ferrari B. Capability of core-sheath polyvinyl alcohol-polycaprolactone emulsion electrospun nanofibrous scaffolds in releasing strontium ranelate for bone regeneration. Biomed Mater 2021; 16:025009. [PMID: 33434897 DOI: 10.1088/1748-605x/abdb07] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Core-sheath nanofibrous scaffolds from polyvinyl alcohol (PVA)-strontium ranelate (SrR)-Polycaprolactone (PCL) were prepared by water in oil electrospinning method. Thus, PCL (the oil phase) was used as the shell part and a mixture of PVA and SrR (the water phase) was inserted in the core. The amounts of SrR was varied from 0 to 15 wt.% Mussel-inspired dopamine-gelatin coating was done on the nanofibrous to improve their hydrophilicity and cellular attachment. The effect of the SrR content on morphology, mechanical, physicochemical, in vitro release behaviors, and biological properties as well as in vivo bone regeneration was investigated. Morphological observations revealed that continuous nanofibers with a core/shell structure were successfully obtained and the fibers diameter increased as the SrR content rose. X-ray diffraction (XRD) analysis revealed that SrR was molecularly distributed in the nanofibers and increasing the amount of the SrR decreased the crystallinity of the nanofibers. Moreover, the SrR release was regulated through the mechanism of Fickian diffusion and it was assumed as fast as possible in the samples with higher SrR content. The mesenchymal stem cell culturing showed improved cell proliferation by adding SrR and accelerating the expression of ALP, Runx2, Col I, and OCN genes. Besides, the SrR-loaded nanofibers improved bone formation of calvarial defects in a rat model as revealed by in vivo investigations.
Collapse
Affiliation(s)
- Seyyed Behnam Abdollahi Boraei
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran. Instituto de Cerámica y Vidrio, CSIC, c/Kelsen 5, 28049 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Rozila I, Azari P, Munirah S, Safwani WKZW, Pingguan-Murphy B, Chua KH. Polycaprolactone-Based Scaffolds Facilitates Osteogenic Differentiation of Human Adipose-Derived Stem Cells in a Co-Culture System. Polymers (Basel) 2021; 13:polym13040597. [PMID: 33671175 PMCID: PMC7922582 DOI: 10.3390/polym13040597] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Stem cells in combination with scaffolds and bioactive molecules have made significant contributions to the regeneration of damaged bone tissues. A co-culture system can be effective in enhancing the proliferation rate and osteogenic differentiation of the stem cells. Hence, the aim of this study was to investigate the osteogenic differentiation of human adipose derived stem cells when co-cultured with human osteoblasts and seeded on polycaprolactone (PCL):hydroxyapatite (HA) scaffold; (2) Methods: Human adipose-derived stem cells (ASC) and human osteoblasts (HOB) were seeded in three different ratios of 1:2, 1:2 and 2:1 in the PCL-HA scaffolds. The osteogenic differentiation ability was evaluated based on cell morphology, proliferation rate, alkaline phosphatase (ALP) activity, calcium deposition and osteogenic genes expression levels using quantitative RT-PCR; (3) Results: The co-cultured of ASC/HOB in ratio 2:1 seeded on the PCL-HA scaffolds showed the most positive osteogenic differentiation as compared to other groups, which resulted in higher ALP activity, calcium deposition and osteogenic genes expression, particularly Runx, ALP and BSP. These genes indicate that the co-cultured ASC/HOB seeded on PCL-HA was at the early stage of osteogenic development; (4) Conclusions: The combination of co-culture system (ASC/HOB) and PCL-HA scaffolds promote osteogenic differentiation and early bone formation.
Collapse
Affiliation(s)
- Ismail Rozila
- Department of Physiology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Cyberjaya, Selangor 63000, Malaysia
| | - Pedram Azari
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Sha’ban Munirah
- Department of Rehabilitation and Physiotherapy, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan 25200, Malaysia;
| | - Wan Kamarul Zaman Wan Safwani
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (W.K.Z.W.S.); (B.P.-M.)
| | - Belinda Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (W.K.Z.W.S.); (B.P.-M.)
| | - Kien Hui Chua
- Department of Physiology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
- Correspondence: ; Tel.: +60-391458613
| |
Collapse
|
16
|
Badekila AK, Kini S, Jaiswal AK. Fabrication techniques of biomimetic scaffolds in three-dimensional cell culture: A review. J Cell Physiol 2021; 236:741-762. [PMID: 32657458 DOI: 10.1002/jcp.29935] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022]
Abstract
In the last four decades, several researchers worldwide have routinely and meticulously exercised cell culture experiments in two-dimensional (2D) platforms. Using traditionally existing 2D models, the therapeutic efficacy of drugs has been inappropriately validated due to the failure in generating the precise therapeutic response. Fortunately, a 3D model addresses the foregoing limitations by recapitulating the in vivo environment. In this context, one has to contemplate the design of an appropriate scaffold for favoring the organization of cell microenvironment. Instituting pertinent model on the platter will pave way for a precise mimicking of in vivo conditions. It is because animal cells in scaffolds oblige spontaneous formation of 3D colonies that molecularly, phenotypically, and histologically resemble the native environment. The 3D culture provides insight into the biochemical aspects of cell-cell communication, plasticity, cell division, cytoskeletal reorganization, signaling mechanisms, differentiation, and cell death. Focusing on these criteria, this paper discusses in detail, the diversification of polymeric scaffolds based on their available resources. The paper also reviews the well-founded and latest techniques of scaffold fabrication, and their applications pertaining to tissue engineering, drug screening, and tumor model development.
Collapse
Affiliation(s)
- Anjana K Badekila
- Nitte University Centre for Science Education and Research, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Sudarshan Kini
- Nitte University Centre for Science Education and Research, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Amit K Jaiswal
- Centre for Biomaterials, Cellular, and Molecular Theranostics, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
17
|
Wu X, Zhang T, Hoff B, Suvarnapathaki S, Lantigua D, McCarthy C, Wu B, Camci‐Unal G. Mineralized Hydrogels Induce Bone Regeneration in Critical Size Cranial Defects. Adv Healthc Mater 2021; 10:e2001101. [PMID: 32940013 DOI: 10.1002/adhm.202001101] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/07/2020] [Indexed: 01/28/2023]
Abstract
Sequential mineralization enables the integration of minerals within the 3D structure of hydrogels. Hydrolyzed collagen-based hydrogels are sequentially mineralized over 10 cycles. One cycle is defined as an incubation period in calcium chloride dihydrate followed by incubation in sodium phosphate dibasic dihydrate. Separate cycles are completed at 30-minute and 24-hour intervals. For the gels mineralized for 30 min and 24 h, the compressive moduli increases from 4.25 to 87.57 kPa and from 4.25 to 125.47 kPa, respectively, as the cycle number increases from 0 to 10. As indicated by X-ray diffraction (XRD) and Fourier transform infrared analysis (FTIR) analyses, the minerals in the scaffolds are mainly hydroxyapatite. In vitro experiments, which measure mechanical properties, porous structure, mineral content, and gene expression are performed to evaluate the physical properties and osteoinductivity of the scaffolds. Real time-quantitative polymerase chain reaction (RT-qPCR) demonstrates 4-10 fold increase in the expression of BMP-7 and osteocalcin. The in vivo subcutaneous implantation demonstrates that the scaffolds are biocompatible and 90% biodegradable. The critical size cranial defects in vivo exhibit nearly complete bone regeneration. Cycle 10 hydrogels mineralized for 24 h have a volume of 59.86 mm3 and a density of 1946.45 HU. These results demonstrate the suitability of sequentially mineralized hydrogel scaffolds for bone repair and regeneration.
Collapse
Affiliation(s)
- Xinchen Wu
- Department of Chemical Engineering University of Massachusetts Lowell Lowell MA 01854 USA
- Biomedical Engineering and Biotechnology Program University of Massachusetts Lowell Lowell MA 01854 USA
| | - Tengfei Zhang
- Department of Neurosurgery Sanbo Brain Hospital Capital Medicine University Beijing 100069 China
| | - Brianna Hoff
- Department of Chemical Engineering University of Massachusetts Lowell Lowell MA 01854 USA
| | - Sanika Suvarnapathaki
- Department of Chemical Engineering University of Massachusetts Lowell Lowell MA 01854 USA
- Biomedical Engineering and Biotechnology Program University of Massachusetts Lowell Lowell MA 01854 USA
| | - Darlin Lantigua
- Department of Chemical Engineering University of Massachusetts Lowell Lowell MA 01854 USA
- Biomedical Engineering and Biotechnology Program University of Massachusetts Lowell Lowell MA 01854 USA
| | - Colleen McCarthy
- Department of Chemical Engineering University of Massachusetts Lowell Lowell MA 01854 USA
| | - Bin Wu
- Department of Neurosurgery Sanbo Brain Hospital Capital Medicine University Beijing 100069 China
| | - Gulden Camci‐Unal
- Department of Chemical Engineering University of Massachusetts Lowell Lowell MA 01854 USA
- Department of Surgery University of Massachusetts Medical School Worcester MA 01605 USA
| |
Collapse
|
18
|
Han X, Xu H, Che L, Sha D, Huang C, Meng T, Song D. Application of Inorganic Nanocomposite Hydrogels in Bone Tissue Engineering. iScience 2020; 23:101845. [PMID: 33305193 PMCID: PMC7711279 DOI: 10.1016/j.isci.2020.101845] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Bone defects caused by trauma and surgery are common clinical problems encountered by orthopedic surgeons. Thus, a hard-textured, natural-like biomaterial that enables encapsulated cells to obtain the much-needed biophysical stimulation and produce functional bone tissue is needed. Incorporating nanomaterials into cell-laden hydrogels is a straightforward tactic for producing tissue engineering structures that integrate perfectly with the body and for tailoring the material characteristics of hydrogels without hindering nutrient exchange with the surroundings. In this review, recent developments in inorganic nanocomposite hydrogels for bone tissue engineering that are of vital importance but have not yet been comprehensively reviewed are summarized.
Collapse
Affiliation(s)
- Xiaying Han
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 New Songjiang Road, Shanghai 200080, China
| | - Houshi Xu
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Lingbin Che
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 New Songjiang Road, Shanghai 200080, China
| | - Dongyong Sha
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Chaojun Huang
- Department of Orthopedics, Shanghai General Hospital, Nanjing Medical University, Shanghai 200080, China
| | - Tong Meng
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 New Songjiang Road, Shanghai 200080, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 New Songjiang Road, Shanghai 200080, China
| |
Collapse
|
19
|
Shariful Islam M, Abdulla-Al-Mamun M, Khan A, Todo M. Excellency of Hydroxyapatite Composite Scaffolds for Bone Tissue Engineering. Biomaterials 2020. [DOI: 10.5772/intechopen.92900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The hydroxyapatite [HAp, Ca10(PO4)6(OH)2] has a variety of applications in bone fillers and replacements due to its excellent bioactivity and osteoconductivity. It comprises the main inorganic component of hard tissues. Among the various approaches, a composite approach using several components like biopolymer, gelatin, collagen, and chitosan in the functionalization of scaffolds with HAp has the prospective to be an engineered biomaterial for bone tissue engineering. HAp composite scaffolds have been developed to obtain a material with different functionalities such as surface reactivity, bioactivity, mechanical strength, and capability of drug or growth factor delivery. Several techniques and processes for the synthesis and fabrication of biocompatible HAp composite scaffolds suitable for bone regeneration are addressed here. Further, this chapter described the excellences of various HAp composite scaffolds used in in vitro and in vivo experiments in bone tissue engineering.
Collapse
|
20
|
Zhou X, Zhou G, Junka R, Chang N, Anwar A, Wang H, Yu X. Fabrication of polylactic acid (PLA)-based porous scaffold through the combination of traditional bio-fabrication and 3D printing technology for bone regeneration. Colloids Surf B Biointerfaces 2020; 197:111420. [PMID: 33113493 DOI: 10.1016/j.colsurfb.2020.111420] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 01/09/2023]
Abstract
Artificial bone grafts possess the advantages of good biodegradability, customizable dimensions, and sufficient mechanical properties, which can promote cell proliferation and differentiation in bone tissue regeneration. 3D printing is a delicate approach that endows the scaffolds with excellent controllability and repeatability when compared with conventional bio-fabrication methods. However, the limitation of printing resolution somehow makes it difficult to prepare bone defect substitution with high porosity and hierarchical construct. In this study, we utilized polylactic acid (PLA) as printing materials and developed a smart strategy to combine 3D printing technology with bio-fabrication methods. A porous planar scaffold was printed and then rolled up into a spiral structure with adjustable pore size and porosity. The topographic features and morphology of the artificial scaffolds were examined through stereomicroscope and SEM, respectively. The porous spiral scaffold presented good mechanical properties in a set of mechanical testing. Later, the human fetal osteoblasts (hFOB) were cultured on the porous spiral scaffold and its control groups for a total of 28 days. The MTS analysis, alkaline phosphatase (ALP) assay, and alizarin red S (ARS) staining were used to analyze the cell proliferation, osteogenic differentiation, and mineral deposition after a certain period of time. The results indicated that compared with the other two scaffolds, the porous spiral scaffold with larger surface area and better interconnections between internal porous networks could significantly improve the spatial cell compartment and promote cell growth and differentiation. The porous spiral scaffold may see versatile applications in large-volume bone defects regeneration.
Collapse
Affiliation(s)
- Xiaqing Zhou
- Department of Biomedical Engineering, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, 07030, United States; Department of Chemistry and Chemical Biology, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
| | - Gan Zhou
- Department of Chemistry and Chemical Biology, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
| | - Radoslaw Junka
- Department of Biomedical Engineering, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
| | - Ningxiao Chang
- Department of Chemistry and Chemical Biology, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
| | - Aneela Anwar
- Department of Biomedical Engineering, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, 07030, United States; Department of Basic Sciences and Humanities, University of Engineering and Technology, New Campus, GT Road, Lahore, 39020, Pakistan
| | - Haoyu Wang
- Department of Biomedical Engineering, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, 07030, United States; Department of Chemistry and Chemical Biology, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
| | - Xiaojun Yu
- Department of Biomedical Engineering, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, 07030, United States.
| |
Collapse
|
21
|
The 3D-Printed Bilayer's Bioactive-Biomaterials Scaffold for Full-Thickness Articular Cartilage Defects Treatment. MATERIALS 2020; 13:ma13153417. [PMID: 32756370 PMCID: PMC7436011 DOI: 10.3390/ma13153417] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 11/22/2022]
Abstract
The full-thickness articular cartilage defect (FTAC) is an abnormally severe grade of articular cartilage (AC) injury. An osteochondral autograft transfer (OAT) is the recommended treatment, but the increasing morbidity rate from osteochondral plug harvesting is a limitation. Thus, the 3D-printed bilayer’s bioactive-biomaterials scaffold is of major interest. Polylactic acid (PLA) and polycaprolactone (PCL) were blended with hydroxyapatite (HA) for the 3D-printed bone layer of the bilayer’s bioactive-biomaterials scaffold (B-BBBS). Meanwhile, the blended PLA/PCL filament was 3D printed and combined with a chitosan (CS)/silk firoin (SF) using a lyophilization technique to fabricate the AC layer of the bilayer’s bioactive-biomaterials scaffold (AC-BBBS). Material characterization and mechanical and biological tests were performed. The fabrication process consists of combining the 3D-printed structure (AC-BBBS and B-BBBS) and a lyophilized porous AC-BBBS. The morphology and printing abilities were investigated, and biological tests were performed. Finite element analysis (FEA) was performed to predict the maximum load that the bilayer’s bioactive-biomaterials scaffold (BBBS) could carry. The presence of HA and CS/SF in the PLA/PCL structure increased cell proliferation. The FEA predicted the load carrying capacity to be up to 663.2 N. All tests indicated that it is possible for BBBS to be used in tissue engineering for AC and bone regeneration in FTAC treatment.
Collapse
|
22
|
Zhao H, Zhang X, Zhou D, Weng Y, Qin W, Pan F, Lv S, Zhao X. Collagen, polycaprolactone and attapulgite composite scaffolds for in vivo bone repair in rabbit models. ACTA ACUST UNITED AC 2020; 15:045022. [PMID: 32224507 DOI: 10.1088/1748-605x/ab843f] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although numerous materials have been explored as bone scaffolds, many of them are limited by their low osteoconductivity and high biodegradability. Therefore, new materials are desired to induce bone cell proliferation and facilitate bone formation. Attapulgite (ATP) is a hydrated silicate that exists in nature as a fibrillar clay mineral and is well known for its large specific surface area, high viscosity, and high absorption capacity, and therefore has the potential to be a new type of bone repair material due to its unique physicochemical properties. In this study, composite scaffolds composed of collagen/polycaprolactone/attapulgite (CPA) or collagen/polycaprolactone (CP) were fabricated through a salt-leaching method. The morphology, composition, microstructure, physical, and mechanical characteristics of the CPA and CP scaffolds were assessed. Cells from the mouse multipotent mesenchymal precursor cell line (D1 cells) were cocultured with the scaffolds, and cell adhesion, proliferation, and gene expression on the CPA and CP scaffolds were analyzed. Adult rabbits with radius defects were used to evaluate the performance of these scaffolds in repairing bone defects over 4-12 weeks. The experimental results showed that the cells demonstrated excellent attachment ability on the CPA scaffolds, as well as remarkable upregulation of the levels of osteoblastic markers such as Runx2, Osterix, collagen 1, osteopontin, and osteocalcin. Furthermore, results from radiography, micro-computed tomography, histological and immunohistochemical analysis demonstrated that abundant new bones were formed on the CPA scaffolds. Ultimately, these results demonstrated that CPA composite scaffolds show excellent potential in bone tissue engineering applications, with the capacity to be used as effective bone regeneration and repair scaffolds in clinical applications.
Collapse
Affiliation(s)
- Hongbin Zhao
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Salifu AA, Obayemi JD, Uzonwanne VO, Soboyejo WO. Mechanical stimulation improves osteogenesis and the mechanical properties of osteoblast-laden RGD-functionalized polycaprolactone/hydroxyapatite scaffolds. J Biomed Mater Res A 2020; 108:2421-2434. [PMID: 32362069 DOI: 10.1002/jbm.a.36993] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/26/2020] [Accepted: 04/04/2020] [Indexed: 11/10/2022]
Abstract
This article presents the results of the combined effects of RGD (arginine-glycine-aspartate) functionalization and mechanical stimulation on osteogenesis that could lead to the development of implantable robust tissue-engineered mineralized constructs. Porous polycaprolactone/hydroxyapatite (PCL/HA) scaffolds are functionalized with RGD-C (arginine-glycine-aspartate-cysteine) peptide. The effects of RGD functionalization are then explored on human fetal osteoblast cell adhesion, proliferation, osteogenic differentiation (alkaline phosphatase activity), extracellular matrix (ECM) production, and mineralization over 28 days. The effects of RGD functionalization followed by mechanical stimulation with a cyclic fluid shear stress of 3.93 mPa in a perfusion bioreactor are also elucidated. The tensile properties (Young's moduli and ultimate tensile strengths) of the cell-laden scaffolds are measured at different stages of cell culture to understand how the mechanical properties of the tissue-engineered structures evolve. RGD functionalization is shown to promote initial cell adhesion, proliferation, alkaline phosphatase (ALP) activity, and ECM production. However, it does not significantly affect mineralization and tensile properties. Mechanical stimulation after RGD functionalization is shown to further improve the ALP activity, ECM production, mineralization, and tensile properties, but not cell proliferation. The results suggest that combined RGD functionalization and mechanical stimulation of cell-laden PCL/HA scaffolds can be used to accelerate the regeneration of robust bioengineered bone structures.
Collapse
Affiliation(s)
- Ali A Salifu
- Materials Science and Engineering Program, Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - John D Obayemi
- Materials Science and Engineering Program, Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Vanessa O Uzonwanne
- Materials Science and Engineering Program, Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Winston O Soboyejo
- Materials Science and Engineering Program, Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
24
|
Zhao H, Tang J, Zhou D, Weng Y, Qin W, Liu C, Lv S, Wang W, Zhao X. Electrospun Icariin-Loaded Core-Shell Collagen, Polycaprolactone, Hydroxyapatite Composite Scaffolds for the Repair of Rabbit Tibia Bone Defects. Int J Nanomedicine 2020; 15:3039-3056. [PMID: 32431500 PMCID: PMC7200251 DOI: 10.2147/ijn.s238800] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Background Electrospinning is a widely used technology that can produce scaffolds with high porosity and surface area for bone regeneration. However, the small pore sizes in electrospun scaffolds constrain cell growth and tissue-ingrowth. In this study, novel drug-loading core-shell scaffolds were fabricated via electrospinning and freeze drying to facilitate the repair of tibia bone defects in rabbit models. Materials and Methods The collagen core scaffolds were freeze-dried containing icariin (ICA)-loaded chitosan microspheres. The shell scaffolds were electrospun using collagen, polycaprolactone and hydroxyapatite materials to form CPH composite scaffolds with the ones containing ICA microspheres named CPHI. The core-shell scaffolds were then cross-linked by genipin. The morphology, microstructure, physical and mechanical properties of the scaffolds were assessed. Rat marrow mesenchymal stem cells from the wistar rat were cultured with the scaffolds. The cell adhesion and proliferation were analysed. Adult rabbit models with tibial plateau defects were used to evaluate the performance of these scaffolds in repairing the bone defects over 4 to 12 weeks. Results The results reveal that the novel drug-loading core-shell scaffolds were successfully fabricated, which showed good physical and chemical properties and appropriate mechanical properties. Furthermore, excellent cells attachment was observed on the CPHI scaffolds. The results from radiography, micro-computed tomography, histological and immunohistochemical analysis demonstrated that abundant new bones were formed on the CPHI scaffolds. Conclusion These new core-shell composite scaffolds have great potential for bone tissue engineering applications and may lead to effective bone regeneration and repair.
Collapse
Affiliation(s)
- Hongbin Zhao
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Junjie Tang
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Dong Zhou
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Yiping Weng
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Wen Qin
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Chun Liu
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Songwei Lv
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, People's Republic of China
| | - Wei Wang
- Medical School, Hexi University, Zhangye 730041, People's Republic of China
| | - Xiubo Zhao
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, People's Republic of China.,Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| |
Collapse
|
25
|
Saxena S, Chang W, Fakhrzadeh A, Murthy NS, Zhang W, Kohn J, Yelick PC. Calcium phosphate enriched synthetic tyrosine-derived polycarbonate - dicalcium phosphate dihydrate polymer scaffolds for enhanced bone regeneration. MATERIALIA 2020; 9:100616. [PMID: 32968719 PMCID: PMC7505226 DOI: 10.1016/j.mtla.2020.100616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Optimal repair of large craniomaxillofacial (CMF) defects caused by trauma or disease requires the development of new, synthetic osteoconductive materials in combination with cell-based therapies, to overcome the limitations of traditionally used bone graft substitutes. In this study, tyrosine-derived polycarbonate, E1001(1k) scaffolds were fabricated to incorporate the osteoinductive coating, Dicalcium phosphate dihydrate (DCPD). The biocompatibility of E1001(1k)-DCPD, E1001(1k)-βTCP and E1001(1k) scaffolds was compared using in vitro culture with human dental pulp stem cells (hDPSCs). We found that the DCPD coating was converted to carbonated hydroxyapatite over time in in vitro culture in Osteogenic Media, while the βTCP did not. hDPSCs exhibited slow initial attachment and proliferation on DCPD E1001(1k) scaffolds, but subsequently improved over time in culture, and promoted osteogenic differentiation. To the best of our knowledge, this study highlights for the first time the effects of Osteogenic Media on phase changes of DCPD, and on DCPD scaffold cytocompatibility with hDPSCs. DCPD showed similar hDPSC biocompatibility and osteoconductivity as compared to βTCP, and osteogenic differentiation of seeded hDPSCs. These studies suggest that E1001(1k)-DCPD scaffolds are a superior tool for craniofacial bone regeneration and provide the foundation for future in vivo testing.
Collapse
Affiliation(s)
- Shruti Saxena
- New Jersey Center for Biomaterials, Rutgers University, Piscataway, New Jersey
| | - Wei Chang
- New Jersey Center for Biomaterials, Rutgers University, Piscataway, New Jersey
| | - Amir Fakhrzadeh
- New Jersey Center for Biomaterials, Rutgers University, Piscataway, New Jersey
| | - N. Sanjeeva Murthy
- New Jersey Center for Biomaterials, Rutgers University, Piscataway, New Jersey
| | - Weibo Zhang
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University School of Dental Medicine, Boston, Massachusetts 02111
| | - Joachim Kohn
- New Jersey Center for Biomaterials, Rutgers University, Piscataway, New Jersey
| | - Pamela C. Yelick
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University School of Dental Medicine, Boston, Massachusetts 02111
| |
Collapse
|
26
|
Jose G, Shalumon K, Liao HT, Kuo CY, Chen JP. Preparation and Characterization of Surface Heat Sintered Nanohydroxyapatite and Nanowhitlockite Embedded Poly (Lactic-co-glycolic Acid) Microsphere Bone Graft Scaffolds: In Vitro and in Vivo Studies. Int J Mol Sci 2020; 21:E528. [PMID: 31947689 PMCID: PMC7013730 DOI: 10.3390/ijms21020528] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 12/12/2022] Open
Abstract
In the context of using bone graft materials to restore and improve the function of damaged bone tissues, macroporous biodegradable composite bone graft scaffolds have osteoinductive properties that allow them to provide a suitable environment for bone regeneration. Hydroxyapatite (HAP) and whitlockite (WLKT) are the two major components of hard tissues such as bone and teeth. Because of their biocompatibility and osteoinductivity, we synthesized HAP (nHAP) and WLKT nanoparticles (nWLKT) by using the chemical precipitation method. The nanoparticles were separately incorporated within poly (lactic-co-glycolic acid) (PLGA) microspheres. Following this, the composite microspheres were converted to macroporous bone grafts with sufficient mechanical strength in pin or screw shape through surface sintering. We characterized physico-chemical and mechanical properties of the nanoparticles and composites. The biocompatibility of the grafts was further tested through in vitro cell adhesion and proliferation studies using rabbit bone marrow stem cells. The ability to promote osteogenic differentiation was tested through alkaline phosphate activity and immunofluorescence staining of bone marker proteins. For in vivo study, the bone pins were implanted in tibia bone defects in rabbits to compare the bone regeneration ability though H&E, Masson's trichrome and immunohistochemical staining. The results revealed similar physico-chemical characteristics and cellular response of PLGA/nHAP and PLGA/nWLKT scaffolds but the latter is associated with higher osteogenic potential towards BMSCs, pointing out the possibility to use this ceramic nanoparticle to prepare a sintered composite microsphere scaffold for potential bone grafts and tissue engineered implants.
Collapse
Affiliation(s)
- Gils Jose
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - K.T. Shalumon
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Han-Tsung Liao
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan
| | - Chang-Yi Kuo
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
27
|
Sultankulov B, Berillo D, Kauanova S, Mikhalovsky S, Mikhalovska L, Saparov A. Composite Cryogel with Polyelectrolyte Complexes for Growth Factor Delivery. Pharmaceutics 2019; 11:pharmaceutics11120650. [PMID: 31817064 PMCID: PMC6955881 DOI: 10.3390/pharmaceutics11120650] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 12/30/2022] Open
Abstract
Macroporous scaffolds composed of chitosan (CHI), hydroxyapatite (HA), heparin (Hep), and polyvinyl alcohol (PVA) were prepared with a glutaraldehyde (GA) cross-linker by cryogelation. Addition of PVA to the reaction mixture slowed down the formation of a polyelectrolyte complex (PEC) between CHI and Hep, which allowed more thorough mixing, and resulted in the development of the homogeneous matrix structure. Freezing of the CHI-HA-GA and PVA-Hep-GA mixture led to the formation of a non-stoichiometric PEC between oppositely charged groups of CHI and Hep, which caused further efficient immobilization of bone morphogenic protein 2 (BMP-2) possible due to electrostatic interactions. It was shown that the obtained cryogel matrix released BMP-2 and supported the differentiation of rat bone marrow mesenchymal stem cells (rat BMSCs) into the osteogenic lineage. Rat BMSCs attached to cryogel loaded with BMP-2 and expressed osteocalcin in vitro. Obtained composite cryogel with PEC may have high potential for bone regeneration and tissue engineering applications.
Collapse
Affiliation(s)
- Bolat Sultankulov
- Department of Chemical Engineering, School of Engineering, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (B.S.); (S.K.)
| | - Dmitriy Berillo
- Department of Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan;
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK;
| | - Sholpan Kauanova
- Department of Chemical Engineering, School of Engineering, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (B.S.); (S.K.)
| | - Sergey Mikhalovsky
- ANAMAD Ltd., Falmer, Brighton BN1 9SB, UK;
- Chuiko Institute of Surface Chemistry, Kyiv 01364, Ukraine
| | - Lyuba Mikhalovska
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK;
| | - Arman Saparov
- School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan
- Correspondence: ; Tel.: +7-7172-706140
| |
Collapse
|
28
|
Choubey R, Chouhan R, Bajpai J, Bajpai AK. Facile Synthesis of Silver Hydroxyapatite (AgHAP) Reinforced Nanocomposites of Poly (styrene)‐Poly (methyl methacrylate) and Study of Their Mechanical and Blood‐Compatible Behavior. ChemistrySelect 2019. [DOI: 10.1002/slct.201902351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Rashmi Choubey
- Bose Memorial Research LaboratoryDepartment of ChemistryGovt. Autonomous Model Science College, Jabalpur, MP India
| | - Raje Chouhan
- Bose Memorial Research LaboratoryDepartment of ChemistryGovt. Autonomous Model Science College, Jabalpur, MP India
| | - Jaya Bajpai
- Bose Memorial Research LaboratoryDepartment of ChemistryGovt. Autonomous Model Science College, Jabalpur, MP India
| | - Anil Kumar Bajpai
- Bose Memorial Research LaboratoryDepartment of ChemistryGovt. Autonomous Model Science College, Jabalpur, MP India
| |
Collapse
|
29
|
Tariverdian T, Behnamghader A, Brouki Milan P, Barzegar-Bafrooei H, Mozafari M. 3D-printed barium strontium titanate-based piezoelectric scaffolds for bone tissue engineering. CERAMICS INTERNATIONAL 2019; 45:14029-14038. [DOI: 10.1016/j.ceramint.2019.04.102] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
|
30
|
Diaz-Gomez L, Kontoyiannis PD, Melchiorri AJ, Mikos AG. Three-Dimensional Printing of Tissue Engineering Scaffolds with Horizontal Pore and Composition Gradients. Tissue Eng Part C Methods 2019; 25:411-420. [PMID: 31169080 PMCID: PMC6657302 DOI: 10.1089/ten.tec.2019.0112] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/24/2019] [Indexed: 12/12/2022] Open
Abstract
IMPACT STATEMENT In this study, we report the development of a novel multimaterial segmented three-dimensional printing methodology to fabricate porous scaffolds containing discrete horizontal gradients of composition and porosity. This methodology is particularly beneficial to preparing porous scaffolds with intricate structures and graded compositions for the regeneration of complex tissues. The technique presented is compatible with many commercially available bioprinters commonly used in biofabrication, and can be adapted to better replicate the architectural and compositional requirements of individual tissues compared with traditional scaffold printing methods.
Collapse
Affiliation(s)
- Luis Diaz-Gomez
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Laboratory, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
| | - Panayiotis D. Kontoyiannis
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Laboratory, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
| | - Anthony J. Melchiorri
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Laboratory, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
| | - Antonios G. Mikos
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Laboratory, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
| |
Collapse
|
31
|
Gupta D, Singh AK, Dravid A, Bellare J. Multiscale Porosity in Compressible Cryogenically 3D Printed Gels for Bone Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2019; 11:20437-20452. [PMID: 31081613 DOI: 10.1021/acsami.9b05460] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Three-dimensional (3D) printing technology has seen several refinements when introduced in the field of medical devices and regenerative medicines. However, it is still a challenge to 3D print gels for building complex constructs as per the desired shape and size. Here, we present a novel method to 3D print gelatin/carboxymethylchitin/hydroxyapatite composite gel constructs of a complex shape. The objective of this study is to fabricate a bioactive gel scaffold with a controlled hierarchical structure. The hierarchy ranges from 3D outer shape to macroporosity to microporosity and rough surface. The fabrication process developed here uses 3D printing in a local cryogenic atmosphere, followed by lyophilization and cross-linking. The gel instantly freezes after extrusion on the cold plate. The cooling action is not limited to the build plate, but the entire gel scaffold is cooled during the 3D printing process. This enables the construction of a stable self-sustaining large-sized 3D complex geometry. Further, lyophilization introduces bulk microporosity into the scaffolds. The outer shape and macroporosity were controlled with the 3D printer, whereas the microporous structure and desirable rough surface morphology were obtained through lyophilization. With cryogenic 3D printing, up to 90% microporosity could be incorporated into the scaffolds. The microporosity and pore size distribution were controlled by changing the cross-linker and total polymer concentration, which resulted in six times increase in surface open pores of size <20 μm on increasing the cross-linker concentration from 25 to 100 mg/mL. The introduction of bulk microporosity was shown to increase swelling by 1.8 times along with a significant increase in human umbilical cord mesenchymal stem cells and Saos-2 cell attachment (2×), proliferation (2.4×), Saos-2 cell alkaline phosphatase level (2×), and mineralization (3×). The scaffolds are spongy in nature in a wet state, thus making them potential implants for bone cavities with a small opening. The application of these cryogenically 3D printed compressible gel scaffolds with multiscale porosity extends to a small- as well as a large-sized open/partially open patient-specific bone defect.
Collapse
Affiliation(s)
| | - Atul Kumar Singh
- Central Research Facility (CRF) , Indian Institute of Technology Delhi , New Delhi 110016 , India
| | - Ashwin Dravid
- Chemical and Biomolecular Engineering , Johns Hopkins University , 323 E 33rd Street , Baltimore , Maryland 21218 , United States
| | | |
Collapse
|
32
|
Kara A, Tamburaci S, Tihminlioglu F, Havitcioglu H. Bioactive fish scale incorporated chitosan biocomposite scaffolds for bone tissue engineering. Int J Biol Macromol 2019; 130:266-279. [PMID: 30797008 DOI: 10.1016/j.ijbiomac.2019.02.067] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 02/01/2019] [Accepted: 02/11/2019] [Indexed: 01/05/2023]
Abstract
Recently, biologically active natural macromolecules have come into prominence to be used as potential materials in scaffold design due to their unique characteristics which can mimic the human tissue structure with their physical and chemical similarity. Among them, fish scale (FS) is a biologically active material with its structural similarity to bone tissue due to including type I collagen and hydroxyapatite and also have distinctive collagen arrangement. In the present study, it is aimed to design a novel composite scaffold with FS incorporation into chitosan (CH) matrix for bone tissue regeneration. Therefore, two biological macromolecules, fish scale and chitosan, were combined to produce bio-composite scaffold. First, FS were decellularized with the chemical method and disrupted physically as microparticles (100 μm), followed by dispersal in CH with ultrasonic homogenisation, CH/FS scaffolds were fabricated by lyophilization technique. Scaffolds were characterized physically, chemically, mechanically, and morphologically. SEM and porosity results showed that CH/FS scaffolds have uniform pore structure showing high porosity. Mechanical properties and degradation rate are enhanced with increasing FS content. In vitro cytotoxicity, proliferation and osteogenic activity of the scaffolds were evaluated with SaOS-2 cell line. CH/FS scaffolds did not show any cytotoxicity effect and the cells were gradually proliferated during culture period. Cell viability results showed that, FS microparticles had a proliferative effect on SaOS-2 cells when compared to control group. ALP activity and biomineralization studies indicated that FS microparticle reinforcement increased osteogenic activity during culture period. As a biological macromolecule with unique characteristics, FS was found as cytocompatible and provided promising effects as reinforcement agents for polymeric scaffolds. In conclusion, fabricated CH/FS bio-composites showed potential for bone tissue engineering applications.
Collapse
Affiliation(s)
- Aylin Kara
- Biotechnology and Bioengineering Graduate Program, Izmir Institute of Technology, Urla, Izmır, Turkey
| | - Sedef Tamburaci
- Biotechnology and Bioengineering Graduate Program, Izmir Institute of Technology, Urla, Izmır, Turkey; Department of Chemical Engineering, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Funda Tihminlioglu
- Department of Chemical Engineering, Izmir Institute of Technology, Urla, Izmir, Turkey.
| | - Hasan Havitcioglu
- Department of Orthopedics and Traumatology, Dokuz Eylul University, Izmır, Turkey
| |
Collapse
|
33
|
Yin S, Zhang W, Zhang Z, Jiang X. Recent Advances in Scaffold Design and Material for Vascularized Tissue-Engineered Bone Regeneration. Adv Healthc Mater 2019; 8:e1801433. [PMID: 30938094 DOI: 10.1002/adhm.201801433] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/24/2019] [Indexed: 12/21/2022]
Abstract
Bone tissue is a highly vascularized tissue and concomitant development of the vascular system and mineralized matrix requires a synergistic interaction between osteogenesis and angioblasts. Several strategies have been applied to achieve vascularized tissue-engineered bone, including the addition of cytokines as well as pre-vascularization strategies and co-culture systems. However, the scaffold is another extremely important component to consider, and development of vascularized bone scaffolds remains one of the greatest challenges for engineering clinically relevant bone substitutes. Here, this review highlights the biomaterial selection, preparation of pre-vascularized scaffolds, composition modification of the scaffold, structural design, and the comprehensive use of the above synergistic modifications of scaffold materials for vascular scaffolds in bone tissue engineering. Moreover, a strategy is proposed for the design of future scaffold structures, in which promoting the regeneration of vascularized bone by regulating the microenvironment should be the main focus. This overview can help illuminate progress in this field and identify the most recently developed scaffolds that show the greatest potential for achieving clinically vascularized bone.
Collapse
Affiliation(s)
- Shi Yin
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No. 639, Manufacturing Bureau Road, Huangpu District, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, No. 639, Manufacturing Bureau Road, Huangpu District, Shanghai, China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No. 639, Manufacturing Bureau Road, Huangpu District, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, No. 639, Manufacturing Bureau Road, Huangpu District, Shanghai, China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Zhiyuan Zhang
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No. 639, Manufacturing Bureau Road, Huangpu District, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, No. 639, Manufacturing Bureau Road, Huangpu District, Shanghai, China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| |
Collapse
|
34
|
Li Y, Liao C, Tjong SC. Synthetic Biodegradable Aliphatic Polyester Nanocomposites Reinforced with Nanohydroxyapatite and/or Graphene Oxide for Bone Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E590. [PMID: 30974820 PMCID: PMC6523566 DOI: 10.3390/nano9040590] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/22/2019] [Accepted: 04/03/2019] [Indexed: 12/27/2022]
Abstract
This paper provides review updates on the current development of bionanocomposites with polymeric matrices consisting of synthetic biodegradable aliphatic polyesters reinforced with nanohydroxyaptite (nHA) and/or graphene oxide (GO) nanofillers for bone tissue engineering applications. Biodegradable aliphatic polyesters include poly(lactic acid) (PLA), polycaprolactone (PCL) and copolymers of PLA-PGA (PLGA). Those bionanocomposites have been explored for making 3D porous scaffolds for the repair of bone defects since nHA and GO enhance their bioactivity and biocompatibility by promoting biomineralization, bone cell adhesion, proliferation and differentiation, thus facilitating new bone tissue formation upon implantation. The incorporation of nHA or GO into aliphatic polyester scaffolds also improves their mechanical strength greatly, especially hybrid GO/nHA nanofilllers. Those mechanically strong nanocomposite scaffolds can support and promote cell attachment for tissue growth. Porous scaffolds fabricated from conventional porogen leaching, and thermally induced phase separation have many drawbacks inducing the use of organic solvents, poor control of pore shape and pore interconnectivity, while electrospinning mats exhibit small pores that limit cell infiltration and tissue ingrowth. Recent advancement of 3D additive manufacturing allows the production of aliphatic polyester nanocomposite scaffolds with precisely controlled pore geometries and large pores for the cell attachment, growth, and differentiation in vitro, and the new bone formation in vivo.
Collapse
Affiliation(s)
- Yuchao Li
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng 252059, China.
| | - Chengzhu Liao
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Sie Chin Tjong
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China.
| |
Collapse
|
35
|
Diaz-Gomez L, Smith BT, Kontoyiannis PD, Bittner SM, Melchiorri AJ, Mikos AG. Multimaterial Segmented Fiber Printing for Gradient Tissue Engineering. Tissue Eng Part C Methods 2019; 25:12-24. [PMID: 30421648 PMCID: PMC6352516 DOI: 10.1089/ten.tec.2018.0307] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
IMPACT STATEMENT This study introduces a segmented three-dimensional printing methodology to create multimaterial porous scaffolds with discrete gradients and controlled distribution of compositions. This methodology can be adapted for the preparation of complex, multimaterial scaffolds with hierarchical structures and mechanical integrity useful in tissue engineering.
Collapse
Affiliation(s)
- Luis Diaz-Gomez
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Lab, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
| | - Brandon T. Smith
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Lab, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
| | - Panayiotis D. Kontoyiannis
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Lab, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
| | - Sean M. Bittner
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Lab, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
| | - Anthony J. Melchiorri
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Lab, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
| | - Antonios G. Mikos
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, Texas
- Biomaterials Lab, Rice University, Houston, Texas
- NIH/NIBIB Center for Engineering Complex Tissues
| |
Collapse
|
36
|
Parmaksiz M, Elçin AE, Elçin YM. Decellularized bovine small intestinal submucosa-PCL/hydroxyapatite-based multilayer composite scaffold for hard tissue repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 94:788-797. [DOI: 10.1016/j.msec.2018.10.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 09/14/2018] [Accepted: 10/02/2018] [Indexed: 12/13/2022]
|
37
|
Wu X, Stroll SI, Lantigua D, Suvarnapathaki S, Camci-Unal G. Eggshell particle-reinforced hydrogels for bone tissue engineering: an orthogonal approach. Biomater Sci 2019; 7:2675-2685. [DOI: 10.1039/c9bm00230h] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Eggshell microparticle-reinforced hydrogels have been fabricated and characterized to obtain mechanically stable and biologically active scaffolds that can direct the differentiation of cells.
Collapse
Affiliation(s)
- Xinchen Wu
- Biomedical Engineering and Biotechnology Program
- University of Massachusetts Lowell
- Lowell
- USA
- Department of Chemical Engineering
| | - Stephanie I. Stroll
- Department of Chemical Engineering
- University of Massachusetts Lowell
- Lowell
- USA
- Department of Biological Sciences
| | - Darlin Lantigua
- Biomedical Engineering and Biotechnology Program
- University of Massachusetts Lowell
- Lowell
- USA
- Department of Chemical Engineering
| | - Sanika Suvarnapathaki
- Biomedical Engineering and Biotechnology Program
- University of Massachusetts Lowell
- Lowell
- USA
- Department of Chemical Engineering
| | - Gulden Camci-Unal
- Department of Chemical Engineering
- University of Massachusetts Lowell
- Lowell
- USA
| |
Collapse
|
38
|
Calcium phosphate compound formed on electrospun chitosan nanofibers using modified simulated body fluid. Polym Bull (Berl) 2018. [DOI: 10.1007/s00289-018-2595-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
39
|
Zhao X, Liu Q, Yang J, Zhang W, Wang Y. Sintering Behavior and Mechanical Properties of Mullite Fibers/Hydroxyapatite Ceramic. MATERIALS 2018; 11:ma11101859. [PMID: 30274239 PMCID: PMC6212928 DOI: 10.3390/ma11101859] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/18/2018] [Accepted: 09/25/2018] [Indexed: 11/24/2022]
Abstract
The effect of fiber content and sintering temperature on sintering behavior and mechanical properties of mullite fibers/hydroxyapatite composites was studied. The composites were fabricated by hydrothermal synthesis and pressureless sintering. The amount of fibers was varied from 5 wt % to 15 wt % through hydrothermal synthesis, mullite fibers and hydroxyapatite composite powders were subsequently sintered at temperatures of 1150, 1250, and 1350 °C. The composites presented a more perturbed structure by increasing fiber content. Moreover, the composites experienced pore coalescence and exhibited a dense microstructure at elevated temperature. X-ray diffraction indicated that the composites underwent various chemical reactions and generated silicate glasses. The generation of silicate glasses increased the driving force of particle rearrangement and decreased the number of pores, which promoted densification of the composites. Densification typically leads to increased hardness and bending strength. The study proposes a densification mechanism and opens new insights into the sintering properties of these materials.
Collapse
Affiliation(s)
- Xueni Zhao
- College of Mechanical and Electrical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Qingyao Liu
- College of Mechanical and Electrical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Jianjun Yang
- College of Mechanical and Electrical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Weigang Zhang
- College of Mechanical and Electrical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Yao Wang
- College of Mechanical and Electrical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| |
Collapse
|
40
|
Sattary M, Rafienia M, Khorasani MT, Salehi H. The effect of collector type on the physical, chemical, and biological properties of polycaprolactone/gelatin/nano-hydroxyapatite electrospun scaffold. J Biomed Mater Res B Appl Biomater 2018; 107:933-950. [PMID: 30199600 DOI: 10.1002/jbm.b.34188] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/22/2018] [Accepted: 06/12/2018] [Indexed: 11/09/2022]
Abstract
Electrospinning is considered a powerful method for the production of fibers in the nanoscale size. Small pore size results in poor cell infiltration, cell migration inhibition into scaffold pores and low oxygen diffusion. Electrospun polycaprolactone/gelatin/nano-hydroxyapatite (PCL/Gel/nHA) scaffolds were deposited into two types of fiber collectors (novel rotating disc and plate) to study fiber morphology, chemical, mechanical, hydrophilic, and biodegradation properties between each other. The proliferation and differentiation of MG-63 cells into the bone phenotype were determined using MTT method, alizarin red staining and alkaline phosphatase (ALP) activity. The rates for disc rotation were 50 and 100 rpm. The pore size measurement results indicated that the fibers produced by the disc rotation collector with speed rate 50 rpm have larger pores as compared to fibers produced by disc rotation at 100 rpm and flat plate collectors. A randomly structure with controlled pore size (38.65 ±0.33 μm) and lower fiber density, as compared to fibers collected by disc rotation with speed rate 100 rpm and flat plate collectors, was obtained. Fibers collected on the rotating disc with speed rate 50 rpm, were more hydrophilic due to larger pore size and therefore, faster infiltration of water into the scaffold and the rate of degradation was higher. These results demonstrate that PCL/Gel/nHA scaffolds made through a rotating disc collector at 50 rpm are more feasible to be used in bone tissue engineering applications due to appropriate pore size and increased adhesion and proliferation of cells, ALP activity and mineral deposits. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 933-950, 2019.
Collapse
Affiliation(s)
- Mansoureh Sattary
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Rafienia
- Biosensor Research Center, Isfahan University of Medical Sciences, 81744*176, Isfahan, Iran
| | - Mohammad Taghi Khorasani
- Department of Biomaterial, Iran Polymer and Petrochemical Institute, PO Box 14965, 159, Tehran, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, 81744*176, Isfahan, Iran
| |
Collapse
|
41
|
Chang W, Shah MB, Lee P, Yu X. Tissue-engineered spiral nerve guidance conduit for peripheral nerve regeneration. Acta Biomater 2018; 73:302-311. [PMID: 29702292 DOI: 10.1016/j.actbio.2018.04.046] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/10/2018] [Accepted: 04/23/2018] [Indexed: 01/09/2023]
Abstract
Recently in peripheral nerve regeneration, preclinical studies have shown that the use of nerve guidance conduits (NGCs) with multiple longitudinally channels and intra-luminal topography enhance the functional outcomes when bridging a nerve gap caused by traumatic injury. These features not only provide guidance cues for regenerating nerve, but also become the essential approaches for developing a novel NGC. In this study, a novel spiral NGC with aligned nanofibers and wrapped with an outer nanofibrous tube was first developed and investigated. Using the common rat sciatic 10-mm nerve defect model, the in vivo study showed that a novel spiral NGC (with and without inner nanofibers) increased the successful rate of nerve regeneration after 6 weeks recovery. Substantial improvements in nerve regeneration were achieved by combining the spiral NGC with inner nanofibers and outer nanofibrous tube, based on the results of walking track analysis, electrophysiology, nerve histological assessment, and gastrocnemius muscle measurement. This demonstrated that the novel spiral NGC with inner aligned nanofibers and wrapped with an outer nanofibrous tube provided a better environment for peripheral nerve regeneration than standard tubular NGCs. Results from this study will benefit for future NGC design to optimize tissue-engineering strategies for peripheral nerve regeneration. STATEMENT OF SIGNIFICANCE We developed a novel spiral nerve guidance conduit (NGC) with coated aligned nanofibers. The spiral structure increases surface area by 4.5 fold relative to a tubular NGC. Furthermore, the aligned nanofibers was coated on the spiral walls, providing cues for guiding neurite extension. Finally, the outside of spiral NGC was wrapped with randomly nanofibers to enhance mechanical strength that can stabilize the spiral NGC. Our nerve histological data have shown that the spiral NGC had 50% more myelinated axons than a tubular structure for nerve regeneration across a 10 mm gap in a rat sciatic nerve. Results from this study can help further optimize tissue engineering strategies for peripheral nerve repair.
Collapse
|
42
|
Zadegan S, Vahidi B, Nourmohammadi J, Haghighipour N. Biocompatibility and bioactivity behaviour of coelectrospun silk fibroin‐hydroxyapatite nanofibres using formic acid. MICRO & NANO LETTERS 2018; 13:709-713. [DOI: 10.1049/mnl.2018.0047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 02/02/2018] [Indexed: 10/06/2024]
Abstract
Appending hydroxyapatite (HAp) to non‐osteogenic silk fibroin (SF) scaffolds is an undeniable fact about the bone tissue engineering. The present research aimed to fabricate the uniform coelectrospun SF‐HAp composites with 1, 3 and 5 wt% of nano‐particles using acid solution to access fibres with favourable mechanical strength simultaneous with the enhanced bone cell compatibility. SEM images displayed that the average diameter of nanofibres increased from 90 ± 12 in SF nanofibres to 227 ± 13 in SF‐5%HAp. Meanwhile, the elastic modulus and breaking strain were equal to 63.4 ± 1.08 MPa and 2.57 ± 0.04, respectively, for SF‐3% HAp which was significantly higher than the SF fibres. Bioactivity property was evaluated after immersion in simulated body fluid by the SEM/EDS analysis. The results, which were confirmed by XRD and FTIR, asserted calcium phosphate deposition after 7 days rather than the SF mats. Finally, the MTT assay, Alkaline Phosphatase (ALP) activity, calcium deposition and cell adhesion approved that the SF‐HAp nano‐composite could raise the cell viability and significantly enhance osteogenic differentiation and mineralisation process by increasing osteoblast cell migration and adhesion to the surface of nanofibres. In brief, it can be concluded that the SF‐HAp nanofibres can be considered as the superlative candidates for application in the bone tissue engineering.
Collapse
Affiliation(s)
- Sara Zadegan
- Division of Biomedical Engineering Department of Life Science Engineering Faculty of New Sciences and Technologies University of Tehran Tehran Iran
| | - Bahman Vahidi
- Division of Biomedical Engineering Department of Life Science Engineering Faculty of New Sciences and Technologies University of Tehran Tehran Iran
| | - Jhamak Nourmohammadi
- Division of Biomedical Engineering Department of Life Science Engineering Faculty of New Sciences and Technologies University of Tehran Tehran Iran
| | | |
Collapse
|
43
|
Wu G, Deng X, Song J, Chen F. Enhanced biological properties of biomimetic apatite fabricated polycaprolactone/chitosan nanofibrous bio-composite for tendon and ligament regeneration. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2018; 178:27-32. [DOI: 10.1016/j.jphotobiol.2017.10.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 09/26/2017] [Accepted: 10/06/2017] [Indexed: 01/27/2023]
|
44
|
Mechanically-competent and cytocompatible polycaprolactone-borophosphosilicate hybrid biomaterials. J Mech Behav Biomed Mater 2017; 75:180-189. [DOI: 10.1016/j.jmbbm.2017.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/30/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022]
|
45
|
Effect of low-intensity pulsed ultrasound on osteogenic human mesenchymal stem cells commitment in a new bone scaffold. J Appl Biomater Funct Mater 2017; 15:e215-e222. [PMID: 28478615 PMCID: PMC6379883 DOI: 10.5301/jabfm.5000342] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose Bone tissue engineering is helpful in finding alternatives to overcome
surgery limitations. Bone growth and repair are under the control of
biochemical and mechanical signals; therefore, in recent years several
approaches to improve bone regeneration have been evaluated. Osteo-inductive
biomaterials, stem cells, specific growth factors and biophysical stimuli
are among those. The aim of the present study was to evaluate if
low-intensity pulsed ultrasound stimulation (LIPUS) treatment would improve
the colonization of an MgHA/Coll hybrid composite scaffold by human
mesenchymal stem cells (hMSCs) and their osteogenic differentiation. LIPUS
stimulation was applied to hMSCs cultured on MgHA/Coll hybrid composite
scaffold in osteogenic medium, mimicking the microenvironment of a bone
fracture. Methods hMSCs were seeded on MgHA/Coll hybrid composite scaffold in an
osteo-inductive medium and exposed to LIPUS treatment for 20 min/day for
different experimental times (7 days, 14 days). The investigation was
focused on (i) the improvement of hMSCs to colonize the MgHA/Coll hybrid
composite scaffold by LIPUS, in terms of cell viability and ultrastructural
analysis; (ii) the activation of MAPK/ERK, osteogenic
(ALPL, COL1A1, BGLAP,
SPP1) and angiogenetic (VEGF, IL8)
pathways, through gene expression and protein release analysis, after LIPUS
stimuli. Results LIPUS exposure improved MgHA/Coll hybrid composite scaffold colonization and
induced in vitro osteogenic differentiation of hMSCs seeded on the
scaffold. Conclusions This work shows that the combined use of new biomimetic osteo-inductive
composite and LIPUS treatment could be a useful therapeutic approach in
order to accelerate bone regeneration pathways.
Collapse
|
46
|
Shi P, Wang Q, Yu C, Fan F, Liu M, Tu M, Lu W, Du M. Hydroxyapatite nanorod and microsphere functionalized with bioactive lactoferrin as a new biomaterial for enhancement bone regeneration. Colloids Surf B Biointerfaces 2017; 155:477-486. [DOI: 10.1016/j.colsurfb.2017.04.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/15/2017] [Accepted: 04/19/2017] [Indexed: 11/25/2022]
|
47
|
Chen L, Wu Z, Zhou Y, Li L, Wang Y, Wang Z, Chen Y, Zhang P. Biomimetic porous collagen/hydroxyapatite scaffold for bone tissue engineering. J Appl Polym Sci 2017. [DOI: 10.1002/app.45271] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Li Chen
- School of Pharmaceutical Sciences; Jilin University; Changchun 130021 People's Republic of China
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry, Chinese Academy of Sciences; Changchun 130022 People's Republic of China
| | - Zhenxu Wu
- School of Pharmaceutical Sciences; Jilin University; Changchun 130021 People's Republic of China
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry, Chinese Academy of Sciences; Changchun 130022 People's Republic of China
| | - Yulai Zhou
- School of Pharmaceutical Sciences; Jilin University; Changchun 130021 People's Republic of China
| | - Linlong Li
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry, Chinese Academy of Sciences; Changchun 130022 People's Republic of China
- University of Chinese Academy of Sciences; Beijing 100039 People's Republic of China
| | - Yu Wang
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry, Chinese Academy of Sciences; Changchun 130022 People's Republic of China
- University of Chinese Academy of Sciences; Beijing 100039 People's Republic of China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry, Chinese Academy of Sciences; Changchun 130022 People's Republic of China
- University of Chinese Academy of Sciences; Beijing 100039 People's Republic of China
| | - Yue Chen
- School of Pharmaceutical Sciences; Jilin University; Changchun 130021 People's Republic of China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry, Chinese Academy of Sciences; Changchun 130022 People's Republic of China
- University of Chinese Academy of Sciences; Beijing 100039 People's Republic of China
| |
Collapse
|
48
|
Salmasi S, Nayyer L, Seifalian AM, Blunn GW. Nanohydroxyapatite Effect on the Degradation, Osteoconduction and Mechanical Properties of Polymeric Bone Tissue Engineered Scaffolds. Open Orthop J 2016; 10:900-919. [PMID: 28217213 PMCID: PMC5299581 DOI: 10.2174/1874325001610010900] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/12/2016] [Accepted: 05/31/2016] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Statistical reports show that every year around the world approximately 15 million bone fractures occur; of which up to 10% fail to heal completely and hence lead to complications of non-union healing. In the past, autografts or allografts were used as the “gold standard” of treating such defects. However, due to various limitations and risks associated with these sources of bone grafts, other avenues have been extensively investigated through which bone tissue engineering; in particular engineering of synthetic bone graft substitutes, has been recognised as a promising alternative to the traditional methods. METHODS A selective literature search was performed. RESULTS Bone tissue engineering offers unlimited supply, eliminated risk of disease transmission and relatively low cost. It could also lead to patient specific design and manufacture of implants, prosthesis and bone related devices. A potentially promising building block for a suitable scaffold is synthetic nanohydroxyapatite incorporated into synthetic polymers. Incorporation of nanohydroxyapatite into synthetic polymers has shown promising bioactivity, osteoconductivity, mechanical properties and degradation profile compared to other techniques previously considered. CONCLUSION Scientific research, through extensive physiochemical characterisation, in vitro and in vivo assessment has brought together the optimum characteristics of nanohydroxyapatite and various types of synthetic polymers in order to develop nanocomposites of suitable nature for bone tissue engineering. The aim of the present article is to review and update various aspects involved in incorporation of synthetic nanohydroxyapatite into synthetic polymers, in terms of their potentials to promote bone growth and regeneration in vitro, in vivo and consequently in clinical applications.
Collapse
Affiliation(s)
- Shima Salmasi
- UCL Division of Surgery and Interventional Science, Centre for Nanotechnology and Regenerative Medicine, University College London, London NW3 2PF, United Kingdom
| | - Leila Nayyer
- UCL Division of Surgery and Interventional Science, Centre for Nanotechnology and Regenerative Medicine, University College London, London NW3 2PF, United Kingdom
| | - Alexander M Seifalian
- UCL Division of Surgery and Interventional Science, Centre for Nanotechnology and Regenerative Medicine, University College London, London NW3 2PF, United Kingdom
| | - Gordon W Blunn
- John Scales Centre for Biomedical Engineering, Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery and Interventional Science, University College London, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, United Kingdom
| |
Collapse
|
49
|
Kaur K, Singh KJ, Anand V, Bhatia G, Kaur R, Kaur M, Nim L, Arora DS. Scaffolds of hydroxyl apatite nanoparticles disseminated in 1, 6-diisocyanatohexane-extended poly(1, 4-butylene succinate)/poly(methyl methacrylate) for bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 71:780-790. [PMID: 27987773 DOI: 10.1016/j.msec.2016.10.055] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/09/2016] [Accepted: 10/23/2016] [Indexed: 11/24/2022]
Abstract
Poly(1, 4-butyl succinate) extended 1, 6-diisocyanatohexane (PBSu-DCH) polymers and Polymethylmethacrylate (PMMA) scaffolds decorated with nano hydroxyl apatite have been prepared and characterized for regeneration of bone in cranio-maxillofacial region. Synthesized scaffolds revealed good response in bone regeneration and excellent cell viability in comparison to commercial available glass plate, which lead to better proliferation of MG-63 cell lines. Additionally, they demonstrate high porosity and excellent water retention ability. Moreover, controlled degradation (in pH=7.4) and sustained drug release in pH (4.5 and 7.4) are advantages of these scaffolds to serve as delivery vehicles for therapeutic drugs. Samples also provide the protection against Escherichia coli and Methicillin Resistant Staphylococcus aureus microorganisms which can be helpful for quick recovery of the patient. In-vitro inflammatory response has been assessed via adsorption of human plasma/serum proteins on the surface of the scaffolds. Results suggest that prepared scaffolds have good bone regeneration ability and provide friendly environment for the cell growth with the additional advantage of protection of the surrounding tissues from microbial infection. With all these features, it is speculated that these scaffolds will have wide utility in the area of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Kulwinder Kaur
- Department of Physics, Guru Nanak Dev University, Amritsar 143005, India
| | - K J Singh
- Department of Physics, Guru Nanak Dev University, Amritsar 143005, India.
| | - Vikas Anand
- Department of Physics, Guru Nanak Dev University, Amritsar 143005, India
| | - Gaurav Bhatia
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar 143005, India
| | - Raminderjit Kaur
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar 143005, India
| | - Manpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar 143005, India
| | - Lovedeep Nim
- Department of Microbiology, Guru Nanak Dev University, Amritsar 143005, India
| | - Daljit Singh Arora
- Department of Microbiology, Guru Nanak Dev University, Amritsar 143005, India
| |
Collapse
|
50
|
Wu CC, Tsai YF, Hsu LH, Chen JP, Sumi S, Yang KC. A self-reinforcing biodegradable implant made of poly(ɛ-caprolactone)/calcium phosphate ceramic composite for craniomaxillofacial fracture fixation. J Craniomaxillofac Surg 2016; 44:1333-41. [DOI: 10.1016/j.jcms.2016.04.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 03/24/2016] [Accepted: 04/15/2016] [Indexed: 11/24/2022] Open
|