1
|
Du J, Zhou T, Peng W. Functional polysaccharide-based hydrogel in bone regeneration: From fundamentals to advanced applications. Carbohydr Polym 2025; 352:123138. [PMID: 39843049 DOI: 10.1016/j.carbpol.2024.123138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/24/2025]
Abstract
Bone regeneration is limited and generally requires external intervention to promote effective repair. Autografts, allografts, and xenografts as traditional methods for addressing bone defects have been widely utilized, their clinical applicability is limited due to their respective disadvantages. Fortunately, functional polysaccharide hydrogels have gained significant attention in bone regeneration due to their exceptional drug-loading capacity, biocompatibility, and ease of chemical modification. They also provide an optimal microenvironment for bone repair and regeneration. This review provides an overview of various functional polysaccharide hydrogels derived from biocompatible materials, focusing on their applications in intelligent delivery systems, bone tissue regeneration, and cartilage defect repair. Particularly, the incorporation of bioactive molecules into the design of functional polysaccharide hydrogels has been shown to significantly enhance bone regeneration. Additionally, this review emphasizes the preparation methods for functional polysaccharide hydrogels and associated the bone healing mechanisms. Finally, the limitations and future prospects of functional polysaccharide hydrogels are thoroughly evaluated.
Collapse
Affiliation(s)
- Jian Du
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100048, China; Hebei North University, Zhangjiakou, 075000, China
| | - Tian Zhou
- Hebei North University, Zhangjiakou, 075000, China
| | - Wei Peng
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100048, China.
| |
Collapse
|
2
|
Zeng Y, Fu BM. Angiogenesis and Microvascular Permeability. Cold Spring Harb Perspect Med 2025; 15:a041163. [PMID: 38692737 PMCID: PMC11694756 DOI: 10.1101/cshperspect.a041163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Angiogenesis, the formation of new blood microvessels, is a necessary physiological process for tissue generation and repair. Sufficient blood supply to the tissue is dependent on microvascular density, while the material exchange between the circulating blood and the surrounding tissue is controlled by microvascular permeability. We thus begin this article by reviewing the key signaling factors, particularly vascular endothelial growth factor (VEGF), which regulates both angiogenesis and microvascular permeability. We then review the role of angiogenesis in tissue growth (bone regeneration) and wound healing. Finally, we review angiogenesis as a pathological process in tumorigenesis, intraplaque hemorrhage, cerebral microhemorrhage, pulmonary fibrosis, and hepatic fibrosis. Since the glycocalyx is important for both angiogenesis and microvascular permeability, we highlight the role of the glycocalyx in regulating the interaction between tumor cells and endothelial cells (ECs) and VEGF-containing exosome release and uptake by tumor-associated ECs, all of which contribute to tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Bingmei M Fu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York 10031, USA
| |
Collapse
|
3
|
Liu G, Zhou L, Li C, Cao J, Zhang Y, Cui S, Liu Y, Xu Y, Zang Y, Yang M, Li Q. Application of Titanium Mesh in the Early Treatment of Flail Chest. Int J Biomater 2024; 2024:8213995. [PMID: 39569041 PMCID: PMC11578650 DOI: 10.1155/2024/8213995] [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: 01/10/2024] [Revised: 07/19/2024] [Accepted: 10/12/2024] [Indexed: 11/22/2024] Open
Abstract
Objective: To investigate the effect of the titanium mesh on flail chest and bone healing from clinical and animal experiments. Methods: Clinical experiment: 24 patients with flail chests in our hospital from January 2020 to January 2023 were prospectively selected and divided into control and titanium mesh groups according to different treatment plans and basic data-matching principles, with 12 cases in each group. The control group was treated with conservative external fixation, and the titanium mesh group was treated with titanium mesh fixation. The clinical efficacy index, visual analog scale and blood gas indexes and hemodynamic indexes of the two groups of patients were recorded. Chest CT and pulmonary function and life quality were examined after operation. Animal experiment: The flail chest sheep were treated conservatively with a titanium mesh, and the expression of bone-healing-related proteins was detected. Results: The mechanical ventilation time, drain indwelling time, ICU observation time, and hospital time in the titanium mesh group were significantly shorter than those in the control group (p < 0.05). The PaO2, CVP, FVC, FEV1, MVV, and life quality of the titanium mesh group were significantly better than those of the control group after operation, and the visual analog scale, PaCO2, CI, ELWI, and the proportions of atelectasis, thoracocyllosis, and consolidation tardive after operation were significantly lower than those of the control group (p < 0.05). The expressions of BMP2, IGF-1, VEGF, and PDGFD in the rib tissue of titanium mesh sheep were higher than those of control sheep at 4 weeks after operation (p < 0.05). Conclusion: Titanium mesh is a safe and effective treatment for flail chest, which can improve pain, blood gas, hemodynamic indexes, and pulmonary function and promote fracture healing.
Collapse
Affiliation(s)
- Gaofeng Liu
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
- Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Li Zhou
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
| | - Chen Li
- Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Junying Cao
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
| | - Yong Zhang
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
| | - Sujuan Cui
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
| | - Yan Liu
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
| | - Yanbin Xu
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
| | - Yongjie Zang
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
| | - Meiming Yang
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
| | - Qingyuan Li
- Department of Cardiothoracic Surgery, The 988th Hospital of PLA Joint Logistics Support, Zhengzhou 450042, Henan Province, China
| |
Collapse
|
4
|
Chen L, Yang J, Cai Z, Huang Y, Xiao P, Wang J, Wang F, Huang W, Cui W, Hu N. Electroactive Biomaterials Regulate the Electrophysiological Microenvironment to Promote Bone and Cartilage Tissue Regeneration. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202314079] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Indexed: 01/06/2025]
Abstract
AbstractThe incidence of large bone and articular cartilage defects caused by traumatic injury is increasing worldwide; the tissue regeneration process for these injuries is lengthy due to limited self‐healing ability. Endogenous bioelectrical phenomenon has been well recognized to play an important role in bone and cartilage homeostasis and regeneration. Studies have reported that electrical stimulation (ES) can effectively regulate various biological processes and holds promise as an external intervention to enhance the synthesis of the extracellular matrix, thereby accelerating the process of bone and cartilage regeneration. Hence, electroactive biomaterials have been considered a biomimetic approach to ensure functional recovery by integrating various physiological signals, including electrical, biochemical, and mechanical signals. This review will discuss the role of endogenous bioelectricity in bone and cartilage tissue, as well as the effects of ES on cellular behaviors. Then, recent advances in electroactive materials and their applications in bone and cartilage tissue regeneration are systematically overviewed, with a focus on their advantages and disadvantages as tissue repair materials and performances in the modulation of cell fate. Finally, the significance of mimicking the electrophysiological microenvironment of target tissue is emphasized and future development challenges of electroactive biomaterials for bone and cartilage repair strategies are proposed.
Collapse
Affiliation(s)
- Li Chen
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Jianye Yang
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Zhengwei Cai
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Yanran Huang
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Pengcheng Xiao
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Juan Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Fan Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Wei Huang
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Wenguo Cui
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Ning Hu
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| |
Collapse
|
5
|
Garcia KR, Beck RCR, Brandalise RN, dos Santos V, Koester LS. Nanocellulose, the Green Biopolymer Trending in Pharmaceuticals: A Patent Review. Pharmaceutics 2024; 16:145. [PMID: 38276515 PMCID: PMC10819157 DOI: 10.3390/pharmaceutics16010145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
The use of nanocellulose in pharmaceutics is a trend that has emerged in recent years. Its inherently good mechanical properties, compared to different materials, such as its high tensile strength, high elastic modulus and high porosity, as well as its renewability and biodegradability are driving nanocellulose's industrial use and innovations. In this sense, this study aims to conduct a search of patents from 2011 to 2023, involving applications of nanocellulose in pharmaceuticals. A patent search was carried out, employing three different patent databases: Patentscope from World Intellectual Property Organization (WIPO); Espacenet; and LENS.ORG. Patents were separated into two main groups, (i) nanocellulose (NC) comprising all its variations and (ii) bacterial nanocellulose (BNC), and classified into five major areas, according to their application. A total of 215 documents was retrieved, of which 179 were referred to the NC group and 36 to the BNC group. The NC group depicted 49.7%, 15.6%, 16.2%, 8.9% and 9.5% of patents as belonging to design and manufacturing, cell culture systems, drug delivery, wound healing and tissue engineering clusters, respectively. The BNC group classified 44.5% of patents as design and manufacturing and 30.6% as drug delivery, as well as 5.6% and 19.4% of patents as wound healing and tissue engineering, respectively. In conclusion, this work compiled and classified patents addressing exclusively the use of nanocellulose in pharmaceuticals, providing information on its current status and trending advancements, considering environmental responsibility and sustainability in materials and products development for a greener upcoming future.
Collapse
Affiliation(s)
- Keth Ribeiro Garcia
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90610-000, Brazil; (K.R.G.); (R.C.R.B.)
| | - Ruy Carlos Ruver Beck
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90610-000, Brazil; (K.R.G.); (R.C.R.B.)
| | - Rosmary Nichele Brandalise
- Programa de Pós-Graduação em Engenharia de Processos e Tecnologias, Universidade de Caxias do Sul (UCS), Caxias do Sul 95070-560, Brazil; (R.N.B.); (V.d.S.)
| | - Venina dos Santos
- Programa de Pós-Graduação em Engenharia de Processos e Tecnologias, Universidade de Caxias do Sul (UCS), Caxias do Sul 95070-560, Brazil; (R.N.B.); (V.d.S.)
| | - Letícia Scherer Koester
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90610-000, Brazil; (K.R.G.); (R.C.R.B.)
| |
Collapse
|
6
|
Nocca G, Arcovito A, Elkasabgy NA, Basha M, Giacon N, Mazzinelli E, Abdel-Maksoud MS, Kamel R. Cellulosic Textiles-An Appealing Trend for Different Pharmaceutical Applications. Pharmaceutics 2023; 15:2738. [PMID: 38140079 PMCID: PMC10747844 DOI: 10.3390/pharmaceutics15122738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Cellulose, the most abundant biopolymer in nature, is derived from various sources. The production of pharmaceutical textiles based on cellulose represents a growing sector. In medicated textiles, textile and pharmaceutical sciences are integrated to develop new healthcare approaches aiming to improve patient compliance. Through the possibility of cellulose functionalization, pharmaceutical textiles can broaden the applications of cellulose in the biomedical field. This narrative review aims to illustrate both the methods of extraction and preparation of cellulose fibers, with a particular focus on nanocellulose, and diverse pharmaceutical applications like tissue restoration and antimicrobial, antiviral, and wound healing applications. Additionally, the merging between fabricated cellulosic textiles with drugs, metal nanoparticles, and plant-derived and synthetic materials are also illustrated. Moreover, new emerging technologies and the use of smart medicated textiles (3D and 4D cellulosic textiles) are not far from those within the review scope. In each section, the review outlines some of the limitations in the use of cellulose textiles, indicating scientific research that provides significant contributions to overcome them. This review also points out the faced challenges and possible solutions in a trial to present an overview on all issues related to the use of cellulose for the production of pharmaceutical textiles.
Collapse
Affiliation(s)
- Giuseppina Nocca
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.N.); (A.A.); (E.M.)
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.N.); (A.A.); (E.M.)
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Nermeen A. Elkasabgy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| | - Mona Basha
- Pharmaceutical Technology Department, National Research Centre, Cairo 12622, Egypt (R.K.)
| | - Noah Giacon
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.N.); (A.A.); (E.M.)
| | - Elena Mazzinelli
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.N.); (A.A.); (E.M.)
| | | | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre, Cairo 12622, Egypt (R.K.)
| |
Collapse
|
7
|
Słota D, Piętak K, Jampilek J, Sobczak-Kupiec A. Polymeric and Composite Carriers of Protein and Non-Protein Biomolecules for Application in Bone Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2235. [PMID: 36984115 PMCID: PMC10059071 DOI: 10.3390/ma16062235] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
Conventional intake of drugs and active substances is most often based on oral intake of an appropriate dose to achieve the desired effect in the affected area or source of pain. In this case, controlling their distribution in the body is difficult, as the substance also reaches other tissues. This phenomenon results in the occurrence of side effects and the need to increase the concentration of the therapeutic substance to ensure it has the desired effect. The scientific field of tissue engineering proposes a solution to this problem, which creates the possibility of designing intelligent systems for delivering active substances precisely to the site of disease conversion. The following review discusses significant current research strategies as well as examples of polymeric and composite carriers for protein and non-protein biomolecules designed for bone tissue regeneration.
Collapse
Affiliation(s)
- Dagmara Słota
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Karina Piętak
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Department of Chemical Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| | - Agnieszka Sobczak-Kupiec
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| |
Collapse
|
8
|
Feng Y, Cölfen H, Xiong R. Organized mineralized cellulose nanostructures for biomedical applications. J Mater Chem B 2023. [PMID: 36892529 DOI: 10.1039/d2tb02611b] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Cellulose is the most abundant naturally-occurring polymer, and possesses a one-dimensional (1D) anisotropic crystalline nanostructure with outstanding mechanical robustness, biocompatibility, renewability and rich surface chemistry in the form of nanocellulose in nature. Such features make cellulose an ideal bio-template for directing the bio-inspired mineralization of inorganic components into hierarchical nanostructures that are promising in biomedical applications. In this review, we will summarize the chemistry and nanostructure characteristics of cellulose and discuss how these favorable characteristics regulate the bio-inspired mineralization process for manufacturing the desired nanostructured bio-composites. We will focus on uncovering the design and manipulation principles of local chemical compositions/constituents and structural arrangement, distribution, dimensions, nanoconfinement and alignment of bio-inspired mineralization over multiple length-scales. In the end, we will underline how these cellulose biomineralized composites benefit biomedical applications. It is expected that this deep understanding of design and fabrication principles will enable construction of outstanding structural and functional cellulose/inorganic composites for more challenging biomedical applications.
Collapse
Affiliation(s)
- Yanhuizhi Feng
- Department of Periodontology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Helmut Cölfen
- Physical Chemistry, Department of Chemistry, University of Konstanz, Universitätsstraße 10, Konstanz, Germany.
| | - Rui Xiong
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu, 610065, China.
| |
Collapse
|
9
|
Janmohammadi M, Nazemi Z, Salehi AOM, Seyfoori A, John JV, Nourbakhsh MS, Akbari M. Cellulose-based composite scaffolds for bone tissue engineering and localized drug delivery. Bioact Mater 2023; 20:137-163. [PMID: 35663339 PMCID: PMC9142858 DOI: 10.1016/j.bioactmat.2022.05.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/27/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022] Open
Abstract
Natural bone constitutes a complex and organized structure of organic and inorganic components with limited ability to regenerate and restore injured tissues, especially in large bone defects. To improve the reconstruction of the damaged bones, tissue engineering has been introduced as a promising alternative approach to the conventional therapeutic methods including surgical interventions using allograft and autograft implants. Bioengineered composite scaffolds consisting of multifunctional biomaterials in combination with the cells and bioactive therapeutic agents have great promise for bone repair and regeneration. Cellulose and its derivatives are renewable and biodegradable natural polymers that have shown promising potential in bone tissue engineering applications. Cellulose-based scaffolds possess numerous advantages attributed to their excellent properties of non-toxicity, biocompatibility, biodegradability, availability through renewable resources, and the low cost of preparation and processing. Furthermore, cellulose and its derivatives have been extensively used for delivering growth factors and antibiotics directly to the site of the impaired bone tissue to promote tissue repair. This review focuses on the various classifications of cellulose-based composite scaffolds utilized in localized bone drug delivery systems and bone regeneration, including cellulose-organic composites, cellulose-inorganic composites, cellulose-organic/inorganic composites. We will also highlight the physicochemical, mechanical, and biological properties of the different cellulose-based scaffolds for bone tissue engineering applications.
Collapse
Affiliation(s)
- Mahsa Janmohammadi
- Faculty of New Sciences and Technologies, Semnan University, Semnan, P.O.Box: 19111-35131, Iran
| | - Zahra Nazemi
- Faculty of New Sciences and Technologies, Semnan University, Semnan, P.O.Box: 19111-35131, Iran
| | | | - Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Johnson V. John
- Terasaki Institute for Biomedical Innovations, Los Angeles, CA, 90050, USA
| | - Mohammad Sadegh Nourbakhsh
- Faculty of Materials and Metallurgical Engineering, Semnan University, Semnan, P.O.Box: 19111-35131, Iran
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Terasaki Institute for Biomedical Innovations, Los Angeles, CA, 90050, USA
- Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100, Gliwice, Poland
| |
Collapse
|
10
|
Park HI, Lee JH, Lee SJ. The comprehensive on-demand 3D bio-printing for composite reconstruction of mandibular defects. Maxillofac Plast Reconstr Surg 2022; 44:31. [PMID: 36195777 PMCID: PMC9532487 DOI: 10.1186/s40902-022-00361-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/25/2022] [Indexed: 11/10/2022] Open
Abstract
Background The mandible is a functional bio-organ that supports facial structures and helps mastication and speaking. Large mandible defects, generally greater than 6-cm segment loss, may require composite tissue reconstruction such as osteocutaneous-vascularized free flap which has a limitation of additional surgery and a functional morbidity at the donor site. A 3D bio-printing technology is recently developed to overcome the limitation in the composite reconstruction of the mandible using osteocutaneous-vascularized free flap. Review Scaffold, cells, and bioactive molecules are essential for a 3D bio-printing. For mandibular reconstruction, materials in a 3D bio-printing require mechanical strength, resilience, and biocompatibility. Recently, an integrated tissue and organ printing system with multiple cartridges are designed and it is capable of printing polymers to reinforce the printed structure, such as hydrogel. Conclusion For successful composite tissue reconstruction of the mandible, biologic considerations and components should be presented with a comprehensive on-demand online platform model of customized approaches.
Collapse
Affiliation(s)
- Han Ick Park
- Department of Oral and Maxillofacial Surgery, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Jee-Ho Lee
- Department of Oral and Maxillofacial Surgery, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea.
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, 27157, USA.
| |
Collapse
|
11
|
Microfluidic 3D Platform to Evaluate Endothelial Progenitor Cell Recruitment by Bioactive Materials. Acta Biomater 2022; 151:264-277. [PMID: 35981686 DOI: 10.1016/j.actbio.2022.08.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 12/30/2022]
Abstract
Most of the conventional in vitro models to test biomaterial-driven vascularization are too simplistic to recapitulate the complex interactions taking place in the actual cell microenvironment, which results in a poor prediction of the in vivo performance of the material. However, during the last decade, cell culture models based on microfluidic technology have allowed attaining unprecedented levels of tissue biomimicry. In this work, we propose a microfluidic-based 3D model to evaluate the effect of bioactive biomaterials capable of releasing signalling cues (such as ions or proteins) in the recruitment of endogenous endothelial progenitor cells, a key step in the vascularization process. The usability of the platform is demonstrated using experimentally-validated finite element models and migration and proliferation studies with rat endothelial progenitor cells (rEPCs) and bone marrow-derived rat mesenchymal stromal cells (BM-rMSCs). As a proof of concept of biomaterial evaluation, the response of rEPCs to an electrospun composite made of polylactic acid with calcium phosphates nanoparticles (PLA+CaP) was compared in a co-culture microenvironment with BM-rMSC to a regular PLA control. Our results show a significantly higher rEPCs migration and the upregulation of several pro-inflammatory and proangiogenic proteins in the case of the PLA+CaP. The effects of osteopontin (OPN) on the rEPCs migratory response were also studied using this platform, suggesting its important role in mediating their recruitment to a calcium-rich microenvironment. This new tool could be applied to screen the capacity of a variety of bioactive scaffolds to induce vascularization and accelerate the preclinical testing of biomaterials. STATEMENT OF SIGNIFICANCE: : For many years researchers have used neovascularization models to evaluate bioactive biomaterials both in vitro, with low predictive results due to their poor biomimicry and minimal control over cell cues such as spatiotemporal biomolecule signaling, and in vivo models, presenting drawbacks such as being highly costly, time-consuming, poor human extrapolation, and ethically controversial. We describe a compact microphysiological platform designed for the evaluation of proangiogenesis in biomaterials through the quantification of the level of sprouting in a mimicked endothelium able to react to gradients of biomaterial-released signals in a fibrin-based extracellular matrix. This model is a useful tool to perform preclinical trustworthy studies in tissue regeneration and to better understand the different elements involved in the complex process of vascularization.
Collapse
|
12
|
A Review of Properties of Nanocellulose, Its Synthesis, and Potential in Biomedical Applications. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12147090] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cellulose is the most venerable and essential natural polymer on the planet and is drawing greater attention in the form of nanocellulose, considered an innovative and influential material in the biomedical field. Because of its exceptional physicochemical characteristics, biodegradability, biocompatibility, and high mechanical strength, nanocellulose attracts considerable scientific attention. Plants, algae, and microorganisms are some of the familiar sources of nanocellulose and are usually grouped as cellulose nanocrystal (CNC), cellulose nanofibril (CNF), and bacterial nanocellulose (BNC). The current review briefly highlights nanocellulose classification and its attractive properties. Further functionalization or chemical modifications enhance the effectiveness and biodegradability of nanocellulose. Nanocellulose-based composites, printing methods, and their potential applications in the biomedical field have also been introduced herein. Finally, the study is summarized with future prospects and challenges associated with the nanocellulose-based materials to promote studies resolving the current issues related to nanocellulose for tissue engineering applications.
Collapse
|
13
|
Patil TV, Patel DK, Dutta SD, Ganguly K, Santra TS, Lim KT. Nanocellulose, a versatile platform: From the delivery of active molecules to tissue engineering applications. Bioact Mater 2022; 9:566-589. [PMID: 34820589 PMCID: PMC8591404 DOI: 10.1016/j.bioactmat.2021.07.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/26/2021] [Accepted: 07/06/2021] [Indexed: 12/13/2022] Open
Abstract
Nanocellulose, a biopolymer, has received wide attention from researchers owing to its superior physicochemical properties, such as high mechanical strength, low density, biodegradability, and biocompatibility. Nanocellulose can be extracted from wide range of sources, including plants, bacteria, and algae. Depending on the extraction process and dimensions (diameter and length), they are categorized into three main types: cellulose nanocrystals (CNCs), cellulose nanofibrils (CNFs), and bacterial nanocellulose (BNC). CNCs are a highly crystalline and needle-like structure, whereas CNFs have both amorphous and crystalline regions in their network. BNC is the purest form of nanocellulose. The nanocellulose properties can be tuned by chemical functionalization, which increases its applicability in biomedical applications. This review highlights the fabrication of different surface-modified nanocellulose to deliver active molecules, such as drugs, proteins, and plasmids. Nanocellulose-mediated delivery of active molecules is profoundly affected by its topographical structure and the interaction between the loaded molecules and nanocellulose. The applications of nanocellulose and its composites in tissue engineering have been discussed. Finally, the review is concluded with further opportunities and challenges in nanocellulose-mediated delivery of active molecules.
Collapse
Affiliation(s)
- Tejal V. Patil
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Dinesh K. Patel
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tuhin Subhra Santra
- Deptarment of Engineering Design, Indian Institute of Technology, Madras, 600036, India
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
14
|
Recent developments of biomaterial scaffolds and regenerative approaches for craniomaxillofacial bone tissue engineering. JOURNAL OF POLYMER RESEARCH 2022. [DOI: 10.1007/s10965-022-02928-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
15
|
Divband B, Aghazadeh M, Al-Qaim ZH, Samiei M, Hussein FH, Shaabani A, Shahi S, Sedghi R. Bioactive chitosan biguanidine-based injectable hydrogels as a novel BMP-2 and VEGF carrier for osteogenesis of dental pulp stem cells. Carbohydr Polym 2021; 273:118589. [PMID: 34560990 DOI: 10.1016/j.carbpol.2021.118589] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/24/2021] [Accepted: 08/17/2021] [Indexed: 12/31/2022]
Abstract
Nowadays, vascularization and mineralization of bone defects is the main bottleneck in the bone regeneration field that is needed to be overcome and developed. Here, we prepared novel in-situ formed injectable hydrogels based on chitosan biguanidine and carboxymethylcellulose loaded with vascular endothelial growth factor (VEGF) and recombinant Bone morphogenetic protein 2 (BMP-2) and studied its influence on osteoblastic differentiation of dental pulp stem cells (DPSCs). The sequential release behavior of the VEGF and BMP-2 from hydrogels adjusted with the pattern of normal human bone growth. MTT assay exhibited that these hydrogels were non-toxic and significantly increased DPSCs proliferation. The Real-time PCR and Western blot analysis on CG11/BMP2-VEGF showed significantly higher gene and protein expression of ALP, COL1α1, and OCN. These results were confirmed by mineralization assay by Alizarin Red staining and Alkaline phosphatase enzyme activity. Based on these evaluations, these hydrogel holds potential as an injectable bone tissue engineering platform.
Collapse
Affiliation(s)
- Baharak Divband
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Inorganic Chemistry Department, Faculty of Chemistry, University of Tabriz, Iran.
| | - Marziyeh Aghazadeh
- Stem Cell Research Center and Oral Medicine, Department of Tabriz, University of Medical Sciences, Iran.
| | - Zahraa Haleem Al-Qaim
- Almustaqbal University College, - Medical Laebroterise, Analytic, Babylon 51005, Iraq
| | - Mohammad Samiei
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Endodontics, Dental School, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Falah H Hussein
- College of Pharmacy, University of Babylon, Babylon 51002, Iraq
| | - Alireza Shaabani
- Department of Polymer and Materials Chemistry, Faculty of Chemistry and Petroleum Sciences, Shahid Beheshti University, Tehran, GC 1983969411, Iran
| | - Shahriar Shahi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Endodontics, Dental School, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Roya Sedghi
- Department of Polymer and Materials Chemistry, Faculty of Chemistry and Petroleum Sciences, Shahid Beheshti University, Tehran, GC 1983969411, Iran.
| |
Collapse
|
16
|
Sun T, Meng C, Ding Q, Yu K, Zhang X, Zhang W, Tian W, Zhang Q, Guo X, Wu B, Xiong Z. In situ bone regeneration with sequential delivery of aptamer and BMP2 from an ECM-based scaffold fabricated by cryogenic free-form extrusion. Bioact Mater 2021; 6:4163-4175. [PMID: 33997500 PMCID: PMC8099605 DOI: 10.1016/j.bioactmat.2021.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
In situ tissue engineering is a powerful strategy for the treatment of bone defects. It could overcome the limitations of traditional bone tissue engineering, which typically involves extensive cell expansion steps, low cell survival rates upon transplantation, and a risk of immuno-rejection. Here, a porous scaffold polycaprolactone (PCL)/decellularized small intestine submucosa (SIS) was fabricated via cryogenic free-form extrusion, followed by surface modification with aptamer and PlGF-2123-144*-fused BMP2 (pBMP2). The two bioactive molecules were delivered sequentially. The aptamer Apt19s, which exhibited binding affinity to bone marrow-derived mesenchymal stem cells (BMSCs), was quickly released, facilitating the mobilization and recruitment of host BMSCs. BMP2 fused with a PlGF-2123-144 peptide, which showed "super-affinity" to the ECM matrix, was released in a slow and sustained manner, inducing BMSC osteogenic differentiation. In vitro results showed that the sequential release of PCL/SIS-pBMP2-Apt19s promoted cell migration, proliferation, alkaline phosphatase activity, and mRNA expression of osteogenesis-related genes. The in vivo results demonstrated that the sequential release system of PCL/SIS-pBMP2-Apt19s evidently increased bone formation in rat calvarial critical-sized defects compared to the sequential release system of PCL/SIS-BMP2-Apt19s. Thus, the novel delivery system shows potential as an ideal alternative for achieving cell-free scaffold-based bone regeneration in situ.
Collapse
Key Words
- 3D, three-dimensional
- Apt19s, aptamer 19s
- Aptamer
- BMD, bone mineral density
- BMP2
- BMP2, bone morphogenic protein 2
- BMSC, bone marrow-derived mesenchymal stem cell
- Bone regeneration in situ
- CLSM, confocal laser scanning microscopy
- CSD, critical-sized calvarial defect
- Cell recruitment
- Controlled delivery
- ECM, decellularized matrix
- FBS, fetal bovine serum
- FDA, US Food and Drug Administration
- FITC, fluorescein isothiocyanate
- FTIR, Fourier transform infrared
- H&E, hematoxylin and eosin
- HA, hydroxyapatite
- PCL, polycaprolactone
- PVDF, polyvinylidene difluoride
- Rh6G, rhodamine 6G
- SIS, small intestine submucosa
- pBMP2, PlGF-2123-144*-fused BMP2
- ssDNA, single-stranded DNA
Collapse
Affiliation(s)
- Tingfang Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chunqing Meng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiuyue Ding
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Keda Yu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xianglin Zhang
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wancheng Zhang
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wenqing Tian
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qi Zhang
- Wuhan Hi-tech Medical Tissue Research Center, Wuhan, 430206, China
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bin Wu
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zekang Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
17
|
Momin M, Mishra V, Gharat S, Omri A. Recent advancements in cellulose-based biomaterials for management of infected wounds. Expert Opin Drug Deliv 2021; 18:1741-1760. [PMID: 34605347 DOI: 10.1080/17425247.2021.1989407] [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: 10/20/2022]
Abstract
INTRODUCTION Chronic wounds are a substantial burden on the healthcare system. Their treatment requires advanced dressings, which can provide a moist wound environment, prevent bacterial infiltration, and act as a drug carrier. Cellulose is biocompatible, biodegradable, and can be functionalized according to specific requirements, which makes it a highly versatile biomaterial. Antimicrobial cellulose dressings are proving to be highly effective against infected wounds. AREAS COVERED This review briefly addresses the mechanism of wound healing and its pathophysiology. It also discusses wound infections, biofilm formation, and progressive emergence of drug-resistant bacteria in chronic wounds and the treatment strategies for such types of infected wounds. It also summarizes the general properties, method of production, and types of cellulose wound dressings. It explores recent studies and advancements regarding the use of cellulose and its derivatives in wound management. EXPERT OPINION Cellulose and its various functionalized derivatives represent a promising choice of wound dressing material. Cellulose-based dressings loaded with antimicrobials are very useful in controlling infection in a chronic wound. Recent studies showing its efficacy against drug-resistant bacteria make it a favorable choice for chronic wound infections. Further research and large-scale clinical trials are required for better clinical evidence of its efficiency.
Collapse
Affiliation(s)
- Munira Momin
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.,SVKM's C B Patel Research Center for Chemistry and Biological Sciences, Mumbai, India
| | - Varsha Mishra
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Sankalp Gharat
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Abdelwahab Omri
- The Novel Drug and Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada
| |
Collapse
|
18
|
Bornert F, Clauss F, Hua G, Idoux-Gillet Y, Keller L, Fernandez De Grado G, Offner D, Smaida R, Wagner Q, Fioretti F, Kuchler-Bopp S, Schulz G, Wenzel W, Gentile L, Risser L, Müller B, Huck O, Benkirane-Jessel N. Mechanistic Illustration: How Newly-Formed Blood Vessels Stopped by the Mineral Blocks of Bone Substitutes Can Be Avoided by Using Innovative Combined Therapeutics. Biomedicines 2021; 9:952. [PMID: 34440156 PMCID: PMC8394928 DOI: 10.3390/biomedicines9080952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/16/2021] [Accepted: 08/01/2021] [Indexed: 12/30/2022] Open
Abstract
One major limitation for the vascularization of bone substitutes used for filling is the presence of mineral blocks. The newly-formed blood vessels are stopped or have to circumvent the mineral blocks, resulting in inefficient delivery of oxygen and nutrients to the implant. This leads to necrosis within the implant and to poor engraftment of the bone substitute. The aim of the present study is to provide a bone substitute currently used in the clinic with suitably guided vascularization properties. This therapeutic hybrid bone filling, containing a mineral and a polymeric component, is fortified with pro-angiogenic smart nano-therapeutics that allow the release of angiogenic molecules. Our data showed that the improved vasculature within the implant promoted new bone formation and that the newly-formed bone swapped the mineral blocks of the bone substitutes much more efficiently than in non-functionalized bone substitutes. Therefore, we demonstrated that our therapeutic bone substitute is an advanced therapeutical medicinal product, with great potential to recuperate and guide vascularization that is stopped by mineral blocks, and can improve the regeneration of critical-sized bone defects. We have also elucidated the mechanism to understand how the newly-formed vessels can no longer encounter mineral blocks and pursue their course of vasculature, giving our advanced therapeutical bone filling great potential to be used in many applications, by combining filling and nano-regenerative medicine that currently fall short because of problems related to the lack of oxygen and nutrients.
Collapse
Affiliation(s)
- Fabien Bornert
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - François Clauss
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Guoqiang Hua
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Ysia Idoux-Gillet
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Laetitia Keller
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Gabriel Fernandez De Grado
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Damien Offner
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Rana Smaida
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Quentin Wagner
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Florence Fioretti
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Sabine Kuchler-Bopp
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Georg Schulz
- Biomaterials Science Center, University of Basel, Gewerbestrasse 14, CH-4123 Allschwil, Switzerland; (G.S.); (B.M.)
| | - Wolfgang Wenzel
- Institute of Nanotechnology, Karlsruhe Institute of Technology, Campus North, Building 640, DE-76131 Karlsruhe, Germany;
| | - Luca Gentile
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Laurent Risser
- Toulouse Institute of Mathematics, UMR 5219 University of Toulouse, CNRS UPS IMT, 31062 Toulouse, France;
| | - Bert Müller
- Biomaterials Science Center, University of Basel, Gewerbestrasse 14, CH-4123 Allschwil, Switzerland; (G.S.); (B.M.)
| | - Olivier Huck
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| |
Collapse
|
19
|
Souza PR, de Oliveira AC, Vilsinski BH, Kipper MJ, Martins AF. Polysaccharide-Based Materials Created by Physical Processes: From Preparation to Biomedical Applications. Pharmaceutics 2021; 13:621. [PMID: 33925380 PMCID: PMC8146878 DOI: 10.3390/pharmaceutics13050621] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Polysaccharide-based materials created by physical processes have received considerable attention for biomedical applications. These structures are often made by associating charged polyelectrolytes in aqueous solutions, avoiding toxic chemistries (crosslinking agents). We review the principal polysaccharides (glycosaminoglycans, marine polysaccharides, and derivatives) containing ionizable groups in their structures and cellulose (neutral polysaccharide). Physical materials with high stability in aqueous media can be developed depending on the selected strategy. We review strategies, including coacervation, ionotropic gelation, electrospinning, layer-by-layer coating, gelation of polymer blends, solvent evaporation, and freezing-thawing methods, that create polysaccharide-based assemblies via in situ (one-step) methods for biomedical applications. We focus on materials used for growth factor (GFs) delivery, scaffolds, antimicrobial coatings, and wound dressings.
Collapse
Affiliation(s)
- Paulo R. Souza
- Group of Polymeric Materials and Composites, Department of Chemistry, State University of Maringá (UEM), Maringá 87020-900, PR, Brazil; (P.R.S.); (A.C.d.O.); (B.H.V.)
| | - Ariel C. de Oliveira
- Group of Polymeric Materials and Composites, Department of Chemistry, State University of Maringá (UEM), Maringá 87020-900, PR, Brazil; (P.R.S.); (A.C.d.O.); (B.H.V.)
- Laboratory of Materials, Macromolecules and Composites, Federal University of Technology—Paraná (UTFPR), Apucarana 86812-460, PR, Brazil
| | - Bruno H. Vilsinski
- Group of Polymeric Materials and Composites, Department of Chemistry, State University of Maringá (UEM), Maringá 87020-900, PR, Brazil; (P.R.S.); (A.C.d.O.); (B.H.V.)
| | - Matt J. Kipper
- Department of Chemical and Biological Engineering, Colorado State University (CSU), Fort Collins, CO 80523, USA
- School of Advanced Materials Discovery, Colorado State University (CSU), Fort Collins, CO 80523, USA
- School of Biomedical Engineering, Colorado State University (CSU), Fort Collins, CO 80523, USA
| | - Alessandro F. Martins
- Group of Polymeric Materials and Composites, Department of Chemistry, State University of Maringá (UEM), Maringá 87020-900, PR, Brazil; (P.R.S.); (A.C.d.O.); (B.H.V.)
- Laboratory of Materials, Macromolecules and Composites, Federal University of Technology—Paraná (UTFPR), Apucarana 86812-460, PR, Brazil
- Department of Chemical and Biological Engineering, Colorado State University (CSU), Fort Collins, CO 80523, USA
| |
Collapse
|
20
|
Abstract
This chapter provides an overview of the growth factors active in bone regeneration and healing. Both normal and impaired bone healing are discussed, with a focus on the spatiotemporal activity of the various growth factors known to be involved in the healing response. The review highlights the activities of most important growth factors impacting bone regeneration, with a particular emphasis on those being pursued for clinical translation or which have already been marketed as components of bone regenerative materials. Current approaches the use of bone grafts in clinical settings of bone repair (including bone grafts) are summarized, and carrier systems (scaffolds) for bone tissue engineering via localized growth factor delivery are reviewed. The chapter concludes with a consideration of how bone repair might be improved in the future.
Collapse
|
21
|
Salehi M, Bastami F, Rezai Rad M, Nokhbatolfoghahaei H, Paknejad Z, Nazeman P, Hassani A, Khojasteh A. Investigation of cell‐free poly lactic acid/nanoclay scaffolds prepared via thermally induced phase separation technique containing hydroxyapatite nanocarriers of erythropoietin for bone tissue engineering applications. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.5120] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Majid Salehi
- Department of Tissue Engineering, School of Medicine Shahroud University of Medical Sciences Shahroud Iran
- Tissue Engineering and Stem Cell Research Center Shahroud University of Medical Sciences Shahroud Iran
| | - Farshid Bastami
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Maryam Rezai Rad
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Zahrasadat Paknejad
- Medical Nanotechnology and Tissue Engineering Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Pantea Nazeman
- Department of Periodontics, School of Dentistry University of Washington Seattle WA USA
| | - Ali Hassani
- Department of Oral and Maxillofacial Surgery and Implant Research Center Islamic Azad University, Tehran Dental Branch Tehran Iran
| | - Arash Khojasteh
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry Shahid Beheshti University of Medical Sciences Tehran Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
22
|
Mansour A, Abu Nada L, El-Hadad AA, Mezour MA, Ersheidat A, Al-Subaie A, Moussa H, Laurenti M, Kaartinen MT, Tamimi F. Biomimetic trace metals improve bone regenerative properties of calcium phosphate bioceramics. J Biomed Mater Res A 2020; 109:666-681. [PMID: 32608184 DOI: 10.1002/jbm.a.37051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 06/07/2020] [Accepted: 06/12/2020] [Indexed: 01/01/2023]
Abstract
The bone regenerative capacity of synthetic calcium phosphates (CaPs) can be enhanced through the enrichment with selected metal trace ions. However, defining the optimal elemental composition required for bone formation is challenging due to many possible concentrations and combinations of these elements. We hypothesized that the ideal elemental composition exists in the inorganic phase of the bone extracellular matrix (ECM). To study our hypothesis, we first obtained natural hydroxyapatite through the calcination of bovine bone, which was then investigated its reactivity with acidic phosphates to produce CaP cements. Bioceramic scaffolds fabricated using these cements were assessed for their composition, properties, and in vivo regenerative performance and compared with controls. We found that natural hydroxyapatite could react with phosphoric acid to produce CaP cements with biomimetic trace metals. These cements present significantly superior in vivo bone regenerative performance compared with cements prepared using synthetic apatite. In summary, this study opens new avenues for further advancements in the field of CaP bone biomaterials by introducing a simple approach to develop biomimetic CaPs. This work also sheds light on the role of the inorganic phase of bone and its composition in defining the regenerative properties of natural bone xenografts.
Collapse
Affiliation(s)
- Alaa Mansour
- Faculty of dentistry, McGill University, Montreal, Canada.,Resident, Periodontics department, School of Dental Medicine, University at Buffalo, Buffalo, New York, USA
| | - Lina Abu Nada
- Faculty of dentistry, McGill University, Montreal, Canada
| | - Amir A El-Hadad
- Faculty of dentistry, McGill University, Montreal, Canada.,Faculty of Science, Al-Azhar University, Cairo, Egypt
| | | | - Ala' Ersheidat
- Faculty of dentistry, McGill University, Montreal, Canada.,Royal Medical Services, Jordanian Armed Forces, Amman, Jordan
| | - Ahmed Al-Subaie
- Faculty of dentistry, McGill University, Montreal, Canada.,College of Dentistry, Imam Abdulrahman Bin Faisal University, Saudi Arabia
| | - Hanan Moussa
- Faculty of dentistry, McGill University, Montreal, Canada.,Faculty of Dentistry, Benghazi University, Benghazi, Libya
| | - Marco Laurenti
- Faculty of Pharmacy, Universidad complutense de Madrid, Spain
| | | | - Faleh Tamimi
- Faculty of dentistry, McGill University, Montreal, Canada.,College of Dental Medicine, Qatar University, Doha, Qatar
| |
Collapse
|
23
|
Rostami F, Tamjid E, Behmanesh M. Drug-eluting PCL/graphene oxide nanocomposite scaffolds for enhanced osteogenic differentiation of mesenchymal stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111102. [PMID: 32600706 DOI: 10.1016/j.msec.2020.111102] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Recently, drug-eluting nanofibrous scaffolds have attracted a great attention to enhance the cell differentiation through biomimicking the extracellular matrix (ECM) in regenerative medicine. In this study, electrospun nanocomposite polycaprolactone (PCL)-based scaffolds containing synthesized graphene oxide (GO) nanosheets and osteogenic drugs, i.e. dexamethasone and simvastatin were fabricated. The physicochemical and surface properties of the scaffolds were investigated through FTIR, wettability, pH, and drug release studies. The cell viability, differentiation, and biomineralization were studied on mesenchymal stem cells (MSCs) by Alamar Blue, alkaline phosphatase (ALP) activity, and Alizarin Red-S staining, respectively. Uniformly distributed GO (thickness < 1 nm) in PCL nanofibers was observed by electron microscopy. It was revealed that the addition of GO and the drugs improved the hydrophilicity, cell viability, and osteogenic differentiation, in addition to pH changes, in comparison with PCL scaffolds. Despite the notable reduction in the cell viability, significant differentiation was revealed by ALP assay on PCL/GO-Dex scaffolds. Noteworthy, a twofold increase in the osteogenic differentiation was observed in comparison with the cells cultured in osteogenic differentiation medium, while a significant biomineralization was observed. The results of this study indicate the synergistic effect of GO and dexamethasone on improving osteogenic differentiation of drug-eluting nanocomposite scaffolds in bone tissue engineering applications.
Collapse
Affiliation(s)
- Fatemeh Rostami
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Elnaz Tamjid
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mehrdad Behmanesh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran; Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
24
|
Rothe R, Hauser S, Neuber C, Laube M, Schulze S, Rammelt S, Pietzsch J. Adjuvant Drug-Assisted Bone Healing: Advances and Challenges in Drug Delivery Approaches. Pharmaceutics 2020; 12:E428. [PMID: 32384753 PMCID: PMC7284517 DOI: 10.3390/pharmaceutics12050428] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/24/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
Bone defects of critical size after compound fractures, infections, or tumor resections are a challenge in treatment. Particularly, this applies to bone defects in patients with impaired bone healing due to frequently occurring metabolic diseases (above all diabetes mellitus and osteoporosis), chronic inflammation, and cancer. Adjuvant therapeutic agents such as recombinant growth factors, lipid mediators, antibiotics, antiphlogistics, and proangiogenics as well as other promising anti-resorptive and anabolic molecules contribute to improving bone healing in these disorders, especially when they are released in a targeted and controlled manner during crucial bone healing phases. In this regard, the development of smart biocompatible and biostable polymers such as implant coatings, scaffolds, or particle-based materials for drug release is crucial. Innovative chemical, physico- and biochemical approaches for controlled tailor-made degradation or the stimulus-responsive release of substances from these materials, and more, are advantageous. In this review, we discuss current developments, progress, but also pitfalls and setbacks of such approaches in supporting or controlling bone healing. The focus is on the critical evaluation of recent preclinical studies investigating different carrier systems, dual- or co-delivery systems as well as triggered- or targeted delivery systems for release of a panoply of drugs.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
| | - Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, 01307 Dresden, Germany; (S.S.); (S.R.)
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, 01307 Dresden, Germany; (S.S.); (S.R.)
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, 01307 Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| |
Collapse
|
25
|
Faruq O, Sayed S, Kim B, Im S, Lee B. A biphasic calcium phosphate ceramic scaffold loaded with oxidized cellulose nanofiber–gelatin hydrogel with immobilized simvastatin drug for osteogenic differentiation. J Biomed Mater Res B Appl Biomater 2020; 108:1229-1238. [DOI: 10.1002/jbm.b.34471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 01/23/2023]
Affiliation(s)
- Omar Faruq
- Department of Regenerative Medicine, College of MedicineSoonchunhyang University Cheonan South Korea
| | - Shithima Sayed
- Department of Regenerative Medicine, College of MedicineSoonchunhyang University Cheonan South Korea
| | - Boram Kim
- Institute of Tissue Regeneration, College of MedicineSoonchunhyang University Cheonan South Korea
| | - Soo‐Bin Im
- Department of Neurosurgery, College of MedicineSoonchunhyang University, Bucheon Hospital Bucheon South Korea
| | - Byong‐Taek Lee
- Department of Regenerative Medicine, College of MedicineSoonchunhyang University Cheonan South Korea
- Institute of Tissue Regeneration, College of MedicineSoonchunhyang University Cheonan South Korea
| |
Collapse
|
26
|
Kuttappan S, Jo JI, Sabu CK, Menon D, Tabata Y, Nair MB. Bioinspired nanocomposite fibrous scaffold mediated delivery of ONO-1301 and BMP2 enhance bone regeneration in critical sized defect. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110591. [DOI: 10.1016/j.msec.2019.110591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 01/08/2023]
|
27
|
Burst, Short, and Sustained Vitamin D 3 Applications Differentially Affect Osteogenic Differentiation of Human Adipose Stem Cells. Int J Mol Sci 2020; 21:ijms21093202. [PMID: 32366057 PMCID: PMC7247321 DOI: 10.3390/ijms21093202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023] Open
Abstract
Incorporation of 1,25(OH)2 vitamin D3 (vitD3) into tissue-engineered scaffolds could aid the healing of critical-sized bone defects. We hypothesize that shorter applications of vitD3 lead to more osteogenic differentiation of mesenchymal stem cells (MSCs) than a sustained application. To test this, release from a scaffold was mimicked by exposing MSCs to exactly controlled vitD3 regimens. Human adipose stem cells (hASCs) were seeded onto calcium phosphate particles, cultured for 20 days, and treated with 124 ng vitD3, either provided during 30 min before seeding ([200 nM]), during the first two days ([100 nM]), or during 20 days ([10 nM]). Alternatively, hASCs were treated for two days with 6.2 ng vitD3 ([10 nM]). hASCs attached to the calcium phosphate particles and were viable (~75%). Cell number was not affected by the various vitD3 applications. VitD3 (124 ng) applied over 20 days increased cellular alkaline phosphatase activity at Days 7 and 20, reduced expression of the early osteogenic marker RUNX2 at Day 20, and strongly upregulated expression of the vitD3 inactivating enzyme CYP24. VitD3 (124 ng) also reduced RUNX2 and increased CYP24 applied at [100 nM] for two days, but not at [200 nM] for 30 min. These results show that 20-day application of vitD3 has more effect on hASCs than the same total amount applied in a shorter time span.
Collapse
|
28
|
Zeng H, Pathak JL, Shi Y, Ran J, Liang L, Yan Q, Wu T, Fan Q, Li M, Bai Y. Indirect selective laser sintering-printed microporous biphasic calcium phosphate scaffold promotes endogenous bone regeneration via activation of ERK1/2 signaling. Biofabrication 2020; 12:025032. [PMID: 32084655 DOI: 10.1088/1758-5090/ab78ed] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The fabrication technique determines the physicochemical and biological properties of scaffolds, including the porosity, mechanical strength, osteoconductivity, and bone regenerative potential. Biphasic calcium phosphate (BCP)-based scaffolds are superior in bone tissue engineering due to their suitable physicochemical and biological properties. We developed an indirect selective laser sintering (SLS) printing strategy to fabricate 3D microporous BCP scaffolds for bone tissue engineering purposes. The green part of the BCP scaffold was fabricated by SLS at a relevant low temperature in the presence of epoxy resin (EP), and the remaining EP was decomposed and eliminated by a subsequent sintering process to obtain the microporous BCP scaffolds. Physicochemical properties, cell adhesion, biocompatibility, in vitro osteogenic potential, and rabbit critical-size cranial bone defect healing potential of the scaffolds were extensively evaluated. This indirect SLS printing eliminated the drawbacks of conventional direct SLS printing at high working temperatures, i.e. wavy deformation of the scaffold, hydroxyapatite decomposition, and conversion of β-tricalcium phosphate (TCP) to α-TCP. Among the scaffolds printed with various binder ratios (by weight) of BCP and EP, the scaffold with 50/50 binder ratio (S4) showed the highest mechanical strength and porosity with the smallest pore size. Scaffold S4 showed the highest effect on osteogenic differentiation of precursor cells in vitro, and this effect was ERK1/2 signaling-dependent. Scaffold S4 robustly promoted precursor cell homing, endogenous bone regeneration, and vascularization in rabbit critical-size cranial defects. In conclusion, BCP scaffolds fabricated by indirect SLS printing maintain the physicochemical properties of BCP and possess the capacity to recruit host precursor cells to the defect site and promote endogenous bone regeneration possibly via the activation of ERK1/2 signaling.
Collapse
Affiliation(s)
- Hao Zeng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Recent Advances of Biphasic Calcium Phosphate Bioceramics for Bone Tissue Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1250:177-188. [PMID: 32601945 DOI: 10.1007/978-981-15-3262-7_12] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biphasic calcium phosphate bioceramics consist of an intimate mixture of hydroxyapatite (HA) and beta-tricalcium phosphate (β-TCP) in varying ratios. Due to their biocompatibility, osteoconductivity, and safety in in vitro, in vivo, and clinical models, they have become promising bone substitute biomaterials and are recommended for use as alternatives for or as additives in bone tissue regeneration in various orthopedic and dental applications. Many studies have demonstrated the potential uses of BCP bioceramics as scaffolds for tissue engineering. Here, we highlight the recent advances in the uses of BCP bioceramics and functionalized BCPs for bone tissue regeneration.
Collapse
|
30
|
Pensa NW, Curry AS, Reddy MS, Bellis SL. Sustained delivery of the angiogenic QK peptide through the use of polyglutamate domains to control peptide release from bone graft materials. J Biomed Mater Res A 2019; 107:2764-2773. [DOI: 10.1002/jbm.a.36779] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 01/19/2023]
Affiliation(s)
- Nicholas W. Pensa
- Department of Biomedical EngineeringUniversity of Alabama at Birmingham Birmingham Alabama
| | - Andrew S. Curry
- Department of Biomedical EngineeringUniversity of Alabama at Birmingham Birmingham Alabama
| | - Michael S. Reddy
- School of DentistryUniversity of California San Francisco California
| | - Susan L. Bellis
- Department of Cell, Developmental, and Integrative BiologyUniversity of Alabama at Birmingham Birmingham Alabama
| |
Collapse
|
31
|
Dual functional approaches for osteogenesis coupled angiogenesis in bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109761. [PMID: 31349418 DOI: 10.1016/j.msec.2019.109761] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/11/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022]
Abstract
Bone fracture healing is a multistep and overlapping process of inflammation, angiogenesis and osteogenesis. It is initiated by inflammation, causing the release of various cytokines and growth factors. It leads to the recruitment of stem cells and formation of vasculature resulting in the functional bone formation. This combined phenomenon is used by bone tissue engineers from past few years to address the problem of vasculature and osteogenic differentiation during bone regeneration. In this review, we have discussed all major studies reporting the dual functioning approach to promote osteogenesis coupled angiogenesis using various scaffolds. These scaffolds are broadly classified into four types based on the nature of their structural and functional components. The functionality of the scaffold is either due to the structural components or the loaded cargo which conducts or induces the coupled functionality. Dual delivery system for osteoinductive and angioinductive factors ensures the co-delivery of two different types of molecules to induce osteogenesis and angiogenesis. Single delivery scaffold for angioinductive and osteoinductive molecule releases single type of molecules which could induce both angiogenesis and osteogenesis. Osteoconductive scaffold consisted of bone constituents releases angioinductive factors. Osteoconductive and angioconductive scaffold composed of components which provide the native substrate features for osteogenesis and angiogenesis. This review article also discusses the studies highlighting the synergism of physico-chemical stimuli as dual functioning feature to enhance angiogenesis and osteogenesis simultaneously. In addition, this article covers one of the least discussed area of the bone regeneration i.e. 'cartilage formation as a median between angiogenesis and osteogenesis'.
Collapse
|
32
|
Zhang Y, Chen H, Zhang T, Zan Y, Ni T, Cao Y, Wang J, Liu M, Pei R. Injectable hydrogels from enzyme-catalyzed crosslinking as BMSCs-laden scaffold for bone repair and regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 96:841-849. [PMID: 30606598 DOI: 10.1016/j.msec.2018.12.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 10/30/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022]
Abstract
Bone-marrow-derived mesenchymal stem cells possess great potential for tissue engineering and regenerative medicine. In the work, an injectable BMSCs-laden hydrogel system was formed by enzyme-catalyzed crosslinking of hyaluronic acid-tyramine and chondroitin sulfate-tyramine in the presence of hydrogen peroxide and horseradish peroxidase, which was used as a 3D scaffold to explore the behavior of the mesenchymal stem cells. Afterward, the gelation rate, mechanical properties, as well as the degradation process of the scaffold were well characterized and optimized. Furthermore, bone morphogenetic protein-2 was encapsulated in the scaffold, which was used to improve the osteogenic properties. The results illustrated that such a BMSCs-laden hydrogel not only offered a proper microenvironment for the adhesion, proliferation and differentiation of mesenchymal stem cells in vitro, but also promoted bone regeneration in vivo. Therefore, this injectable BMSCs-laden hydrogel may serve as an efficient 3D scaffold for bone repair and regeneration.
Collapse
Affiliation(s)
- Yajie Zhang
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Hong Chen
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Tingting Zhang
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yue Zan
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Tianyu Ni
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; Nano Science and Technology Institute, University of Science and Technology of China, Suzhou 215123, China
| | - Yi Cao
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jine Wang
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Min Liu
- Institute for Interdisciplinary Research, Jianghan University, Wuhan 430056, China.
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
33
|
Cao L, Kong X, Lin S, Zhang S, Wang J, Liu C, Jiang X. Synergistic effects of dual growth factor delivery from composite hydrogels incorporating 2-N,6-O-sulphated chitosan on bone regeneration. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S1-S17. [PMID: 30231646 DOI: 10.1080/21691401.2018.1488721] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A promising strategy to accelerate bone generation is to deliver a combination of certain growth factors to the integration site via a controlled spatial and temporal delivery mode. Here, a composite hydrogel incorporating poly(lactide-co-glycolide) (PLGA) microspheres was accordingly prepared to load and deliver the osteogenic rhBMP-2 and angiogenic rhVEGF165 in the required manner. In addition, 2-N,6-O-sulphated chitosan (26SCS), which is a synergetic factor of growth factors, was incorporated in the composite hydrogel as well. The system showed a similar release behaviour of the two growth factors regardless of 26SCS inclusion. RhBMP-2 loaded in PLGA microspheres showed a sustained release over a period of 2 weeks, whereas rhVEGF165 loaded in hydrogel eluted almost completely from the hydrogel over the first 16 days. Both growth factors retained their efficacy, as quantified with relevant in vitro assays. Moreover, an enhanced cell response was achieved upon the delivery of dual growth factors, compared to that obtained with a single factor. Furthermore, in the presence of 26SCS, the system revealed significantly upregulated alkaline phosphatase activity, human umbilical vein endothelial cell proliferation, sprouting, nitric oxide secretion, and angiogenic gene expression. This study highlighted that the composite hydrogel incorporated with 26SCS appears to constitute a promising approach to deliver multiple growth factors. From our findings, we could also conclude that rhBMP-2 can promote angiogenesis and that the mechanism is worthy of further study in subsequent research.
Collapse
Affiliation(s)
- Lingyan Cao
- a Department of Prosthodontics , Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, PR China.,b Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , National Clinical Research Center of Stomatology , Shanghai , PR China
| | - Xiangjun Kong
- c Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China
| | - Shuxian Lin
- a Department of Prosthodontics , Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, PR China.,b Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , National Clinical Research Center of Stomatology , Shanghai , PR China
| | - Shuang Zhang
- c Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China
| | - Jing Wang
- c Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China
| | - Changsheng Liu
- c Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China.,d Key Laboratory for Ultrafine Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China
| | - Xinquan Jiang
- a Department of Prosthodontics , Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, PR China.,b Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , National Clinical Research Center of Stomatology , Shanghai , PR China
| |
Collapse
|
34
|
Kuttappan S, Mathew D, Jo JI, Tanaka R, Menon D, Ishimoto T, Nakano T, Nair SV, Nair MB, Tabata Y. Dual release of growth factor from nanocomposite fibrous scaffold promotes vascularisation and bone regeneration in rat critical sized calvarial defect. Acta Biomater 2018; 78:36-47. [PMID: 30067947 DOI: 10.1016/j.actbio.2018.07.050] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/02/2018] [Accepted: 07/27/2018] [Indexed: 11/27/2022]
Abstract
A promising strategy for augmenting bone formation involves the local delivery of multiple osteoinductive and vasculogenic growth factors. However, success depends on sustained growth factor release and its appropriate combination to induce stem cells and osteogenic cells at the bony site. Herein, we have developed a nanocomposite fibrous scaffold loaded with fibroblast growth factor 2 (FGF2), vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP2) and its ability to promote vascularisation and bone regeneration in critical sized calvarial defect was compared to the scaffold with VEGF + BMP2 and FGF2 + BMP2. Simple loading of growth factors on the scaffold could provide a differential release pattern, both in vitro and in vivo (VEGF release for 1 week where as BMP2 and FGF2 release for 3 weeks). Among all the groups, dual growth factor loaded scaffold (VEGF + BMP2 & FGF2 + BMP2) enhanced vascularisation and new bone formation, but there was no difference between FGF2 and VEGF loaded scaffolds although its release pattern was different. FGF2 mainly promoted stem cell migration, whereas VEGF augmented new blood vessel formation at the defect site. This study suggests that biomimetic nanocomposite scaffold is a promising growth factor delivery vehicle to improve bone regeneration in critical sized bone defects. STATEMENT OF SIGNIFICANCE Many studies have shown the effect of growth factors like VEGF-BMP2 or FGF2-BMP2 in enhancing bone formation in critical sized defects, but there are no reports that demonstrate the direct comparison of VEGF-BMP2 and FGF2-BMP2. In this study, we have developed a nanocomposite fibrous scaffold that could differentially release growth factors like VEGF, BMP2 and FGF2 (VEGF release for 1 week where as BMP2 and FGF2 release for 3 weeks), which in turn promoted neovascularisation and new bone formation in critical sized defects. There was no difference in vascularisation and bone formation induced by VEGF + BMP2 or FGF2 + BMP2. The growth factor was loaded in a simple manner, which would ensure ease of use for the end-user, especially for the surgeons treating a patient in an operating room.
Collapse
|
35
|
Zhang B, Li H, He L, Han Z, Zhou T, Zhi W, Lu X, Lu X, Weng J. Surface-decorated hydroxyapatite scaffold with on-demand delivery of dexamethasone and stromal cell derived factor-1 for enhanced osteogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 89:355-370. [DOI: 10.1016/j.msec.2018.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 03/17/2018] [Accepted: 04/09/2018] [Indexed: 12/17/2022]
|
36
|
Wong SA, Rivera KO, Miclau T, Alsberg E, Marcucio RS, Bahney CS. Microenvironmental Regulation of Chondrocyte Plasticity in Endochondral Repair-A New Frontier for Developmental Engineering. Front Bioeng Biotechnol 2018; 6:58. [PMID: 29868574 PMCID: PMC5962790 DOI: 10.3389/fbioe.2018.00058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/23/2018] [Indexed: 12/31/2022] Open
Abstract
The majority of fractures heal through the process of endochondral ossification, in which a cartilage intermediate forms between the fractured bone ends and is gradually replaced with bone. Recent studies have provided genetic evidence demonstrating that a significant portion of callus chondrocytes transform into osteoblasts that derive the new bone. This evidence has opened a new field of research aimed at identifying the regulatory mechanisms that govern chondrocyte transformation in the hope of developing improved fracture therapies. In this article, we review known and candidate molecular pathways that may stimulate chondrocyte-to-osteoblast transformation during endochondral fracture repair. We also examine additional extrinsic factors that may play a role in modulating chondrocyte and osteoblast fate during fracture healing such as angiogenesis and mineralization of the extracellular matrix. Taken together the mechanisms reviewed here demonstrate the promising potential of using developmental engineering to design therapeutic approaches that activate endogenous healing pathways to stimulate fracture repair.
Collapse
Affiliation(s)
- Sarah A Wong
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,School of Dentistry, University of California, San Francisco, San Francisco, CA, United States
| | - Kevin O Rivera
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,School of Dentistry, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Eben Alsberg
- Department of Orthopaedic Surgery and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Ralph S Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,School of Dentistry, University of California, San Francisco, San Francisco, CA, United States
| | - Chelsea S Bahney
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
37
|
Wang X, Ding Z, Wang C, Chen X, Xu H, Lu Q, Kaplan DL. Bioactive Silk Hydrogels with Tunable Mechanical Properties. J Mater Chem B 2018; 6:2739-2746. [PMID: 30345058 PMCID: PMC6191054 DOI: 10.1039/c8tb00607e] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Developing bioactive hydrogels with potential to guide the differentiation behavior of stem cells has become increasingly important in the biomaterials field. Here, silk hydrogels with tunable mechanical properties were developed by introducing inert silk fibroin nanofibers (SNF) within an enzyme crosslinked system of regenerated silk fibroin (RSF). After the crosslinking reaction of RSF, the inert SNF was embedded into the RSF hydrogel matrix, resulting in improved mechanical properties. Tunable stiffness in the range of 9-60 KPa was achieved by adjusting the amount of the added NSF, significantly higher than SNF-free hydrogels formed under same conditions (about 1 KPa). In addition, the proliferation of rat bone marrow derived mesenchymal stem cells cultured on the composite hydrogels and differentiated into endothelial cells, myoblast and osteoblast cells was improved, putatively due to the control of stiffness of the hydrogels. Bioactive and tunable silk-based hydrogels were prepared via a composite SNF and crosslinked RSF system, providing a new strategy to design silk biomaterials with tunable mechanical and biological performance.
Collapse
Affiliation(s)
- Xue Wang
- Department of Dermatology and Dermatologic Surgery, Shanghai Ninth People's Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, P. R. China
| | - Zhaozhao Ding
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science, Soochow University, Suzhou 215123, P. R. China
| | - Chen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, P. R. China
| | - Xiangdong Chen
- Department of Dermatology and Dermatologic Surgery, Shanghai Ninth People's Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, P. R. China
| | - Hui Xu
- Department of Dermatology and Dermatologic Surgery, Shanghai Ninth People's Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, P. R. China
| | - Qiang Lu
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science, Soochow University, Suzhou 215123, P. R. China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
38
|
Wei F, Zhou Y, Wang J, Liu C, Xiao Y. The Immunomodulatory Role of BMP-2 on Macrophages to Accelerate Osteogenesis. Tissue Eng Part A 2018; 24:584-594. [DOI: 10.1089/ten.tea.2017.0232] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Fei Wei
- The Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| | - Yinghong Zhou
- The Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| | - Jing Wang
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Changsheng Liu
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Yin Xiao
- The Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
39
|
Imamura K, Tachi K, Takayama T, Shohara R, Kasai H, Dai J, Yamano S. Released fibroblast growth factor18 from a collagen membrane induces osteoblastic activity involved with downregulation of miR-133a and miR-135a. J Biomater Appl 2018; 32:1382-1391. [PMID: 29544382 DOI: 10.1177/0885328218763318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We have developed a unique delivery system of growth factors using collagen membranes (CMs) to induce bone regeneration. We hypothesized that fibroblast growth factor18 (FGF-18), a pleiotropic protein that stimulates proliferation in several tissues, can be a good candidate to use our delivery system for bone regeneration. Cell viability, cell proliferation, alkaline phosphatase activity, mineralization, and marker gene expression of osteoblastic differentiation were evaluated after mouse preosteoblasts were cultured with a CM containing FGF-18, a CM containing platelet-derived growth factor, or a CM alone. Furthermore, expression of microRNA, especially miR-133a and miR-135a involving inhibition of osteogenic factors, was measured in preosteoblasts with CM/FGF-18 or CM alone. A sustained release of FGF-18 from the CM was observed over 21 days. CM/FGF-18 significantly promoted cell proliferation, alkaline phosphatase activity, and mineralization compared to CM alone. Gene expression of type I collagen, runt-related transcription factor 2, osteocalcin, Smad5, and osteopontin was significantly upregulated in CM/FGF-18 compared to CM alone, and similar to CM/platelet-derived growth factor. Additionally, CM/FGF-18 downregulated expression of miR-133a and miR-135a. These results suggested that released FGF-18 from a CM promotes osteoblastic activity involved with downregulation of miR-133a and miR-135a.
Collapse
Affiliation(s)
- Kentaro Imamura
- 1 Department of Prosthodontics, 70241 New York University College of Dentistry , NY, USA.,2 Department of Periodontology, Tokyo Dental College, Tokyo, Japan
| | - Keita Tachi
- 1 Department of Prosthodontics, 70241 New York University College of Dentistry , NY, USA
| | - Tadahiro Takayama
- 3 Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan
| | - Ryutaro Shohara
- 1 Department of Prosthodontics, 70241 New York University College of Dentistry , NY, USA
| | - Hironori Kasai
- 1 Department of Prosthodontics, 70241 New York University College of Dentistry , NY, USA
| | - Jisen Dai
- 4 Mouse Genotyping Core, New York University Langone Medical Center, NY, USA
| | - Seiichi Yamano
- 1 Department of Prosthodontics, 70241 New York University College of Dentistry , NY, USA
| |
Collapse
|
40
|
Kim B, Ventura R, Lee BT. Functionalization of porous BCP scaffold by generating cell-derived extracellular matrix from rat bone marrow stem cells culture for bone tissue engineering. J Tissue Eng Regen Med 2017; 12:e1256-e1267. [PMID: 28752541 DOI: 10.1002/term.2529] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 05/24/2017] [Accepted: 07/24/2017] [Indexed: 12/24/2022]
Abstract
The potential of decellularized cell-derived extracellular matrix (ECM) deposited on biphasic calcium phosphate (BCP) scaffold for bone tissue engineering was investigated. Rat derived bone marrow mesenchymal stem cells were cultured on porous BCP scaffolds for 3 weeks and decellularized with two different methods (freeze-thaw [F/T] or sodium dodecyl sulfate [SDS]). The decellularized ECM deposited scaffolds (dECM-BCP) were characterized through scanning electron microscopy, energy dispersive X-ray spectrometer, and confocal microscopy. The efficiency of decellularization was evaluated by quantifying remaining DNA, sulfated glycosaminoglycans, and collagens. Results revealed that F/T method was more effective procedure for removing cellular components of cultured cells (95.21% DNA reduction) than SDS treatment (92.49%). Although significant loss of collagen was observed after decellularization with both F/T (56.68%) and SDS (70.87%) methods, F/T treated sample showed higher retaining amount of sulfated glycosaminoglycans content (75.64%) than SDS (33.28%). In addition, we investigated the cell biocompatibility and osteogenic effect of dECM-BCP scaffolds using preosteoblasts (MC3T3-E1). Compared to bare BCP scaffolds, dECM-BCP_F/T scaffolds showed improved cell attachment and proliferation based on immunofluorescence staining and water soluble tetrazolium salts assay (p < .001). Moreover, dECM-BCP scaffolds showed increased osteoblastic differentiation of newly seeded preosteoblasts by up-regulating three types of osteoblastic genes (osteopontin, alkaline phosphatase, and bone morphogenic protein-2). This study demonstrated that functionalization of BCP scaffold using cell-derived ECM could be useful for improving the bioactivity of materials and providing suitable microenvironment, especially for osteogenesis. Further study is needed to determine the potential of dECM-BCP scaffold for bone formation and regeneration in vivo.
Collapse
Affiliation(s)
- Boram Kim
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Reiza Ventura
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Byong-Taek Lee
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, Korea.,Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Korea
| |
Collapse
|
41
|
Liao H, Zhong Z, Liu Z, Li L, Ling Z, Zou X. Bone mesenchymal stem cells co-expressing VEGF and BMP-6 genes to combat avascular necrosis of the femoral head. Exp Ther Med 2017; 15:954-962. [PMID: 29399103 PMCID: PMC5772743 DOI: 10.3892/etm.2017.5455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/03/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate the potential of bone mesenchymal stem cells (BMSCs) treated with a combination of vascular endothelial growth factor (VEGF) and bone morphogenetic protein-6 (BMP-6) genes for the treatment of avascular necrosis of the femoral head (ANFH). Rat BMSCs were isolated and purified using a density gradient centrifugation method. The purity and characteristics of the BMSCs were detected by cell surface antigens identification using flow cytometry. The experimental groups were administered with one of the following adeno-associated virus (AAV) vector constructs: AAV-green fluorescent protein (AAV-GFP), AAV-BMP-6, AAV-VEGF or AAV-VEGF-BMP-6. The expression of VEGF and BMP-6 was detected by reverse transcription-quantitative polymerase chain reaction, western blotting and ELISA assays. The effects of VEGF and BMP-6 on BMSCs were evaluated by angiogenic and osteogenic assays. The transfected BMSCs were combined with a biomimetic synthetic scaffold poly lactide-co-glycolide (PLAGA) and they were then subcutaneously implanted into nude mice. After four weeks, the implants were analyzed with histology and subsequent immunostaining to evaluate the effects of BMSCs on blood vessel and bone formation in vivo. In the AAV-VEGF-BMP-6 group, the expression levels of VEGF and BMP-6 were significantly increased and human umbilical vein endothelial cells tube formation was significantly enhanced compared with other groups. Capillaries and bone formation in the AAV-VEGF-BMP-6 group was significantly higher compared with the other groups. The results of the present study suggest that BMSCs expressing both VEGF and BMP-6 induce an increase in blood vessels and bone formation, which provides theoretical support for ANFH gene therapy.
Collapse
Affiliation(s)
- Hongxing Liao
- Department of Orthopedics, Meizhou People's Hospital, Meizhou, Guangdong 514000, P.R. China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhixiong Zhong
- Department of Cardiovascular Medicine, Meizhou People's Hospital, Meizhou, Guangdong 514000, P.R. China
| | - Zhanliang Liu
- Department of Orthopedics, Meizhou People's Hospital, Meizhou, Guangdong 514000, P.R. China
| | - Liangping Li
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zemin Ling
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
42
|
Hasan ML, Taz M, Lee BT. Effects of platelet-rich plasma on biological activity and bone regeneration of brushite-based calcium phosphate cement. J Biomed Mater Res B Appl Biomater 2017; 106:2316-2326. [DOI: 10.1002/jbm.b.34036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 09/12/2017] [Accepted: 10/13/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Md Lemon Hasan
- Department of Regenerative Medicine; College of Medicine, Soonchunhyang University, Cheonan-City; ChungCheongNam-Do South Korea
| | - Mirana Taz
- Department of Regenerative Medicine; College of Medicine, Soonchunhyang University, Cheonan-City; ChungCheongNam-Do South Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine; College of Medicine, Soonchunhyang University, Cheonan-City; ChungCheongNam-Do South Korea
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan-City; ChungCheongNam-Do South Korea
| |
Collapse
|
43
|
Ou Z, Cheng Q, Chen Y, Chen T, Rong X, Long F, Zhang X, Liang Q, Feng Z. Chemical characterization of wound ointment (WO) and its effects on fracture repair: a rabbit model. Chin Med 2017; 12:31. [PMID: 29093746 PMCID: PMC5661936 DOI: 10.1186/s13020-017-0152-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
Background Wound ointment (WO), a kind of Chinese medicine, can significantly promote fracture healing. The study aimed at analyzing the chemical composition and the effects of WO on fracture of rabbits and tried to explore the corresponding molecular mechanism in cytokine. Methods The qualitative and quantitative analysis of WO was conducted by liquid chromatography-mass spectrometry (LC–MS). Fifty-four Zealand mature male rabbits were randomly divided into 3 groups: Control group, Yunnan Baiyao (YB) group and WO group. All the rabbits suffered a fracture of right radius and were then stabilized with an external fixator. Treated with different methods, fracture healing was observed. The bone specimens were subjected to radiograph, immunohistochemistry (IHC) analysis, hematoxylin–eosin staining (HE), western blot and enzyme linked immunosorbent assay (ELISA). Results A total of 12 active compositions were detected by LC–MS. Radiographs showed a considerably better bone healing and remodeling of the fracture in WO group. HE experiments showed that a large number of osteoclasts appeared in the early stage when treated with WO. In immunohistochemistry (IHC), western blot and ELISA test, significant increases in vascular endothelial growth factor (VEGF) expression were observed in WO group compared with other two groups. Conclusions Wound ointment contained active compositions which efficiently promoted fracture healing through increasing the expression of VEGF. Trial Registration Not applicable Electronic supplementary material The online version of this article (doi:10.1186/s13020-017-0152-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhixue Ou
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Qi Cheng
- Department of Traumatic Orthopedics, Daye Hospital of Traditional Chinese Medicine, Daye, 435100 Hubei China
| | - Yueping Chen
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Tao Chen
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Xiangbin Rong
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Feipan Long
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Xiaoyun Zhang
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Qinghua Liang
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Zhe Feng
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| |
Collapse
|
44
|
Thrivikraman G, Athirasala A, Twohig C, Boda SK, Bertassoni LE. Biomaterials for Craniofacial Bone Regeneration. Dent Clin North Am 2017; 61:835-856. [PMID: 28886771 PMCID: PMC5663293 DOI: 10.1016/j.cden.2017.06.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Functional reconstruction of craniofacial defects is a major clinical challenge in craniofacial sciences. The advent of biomaterials is a potential alternative to standard autologous/allogenic grafting procedures to achieve clinically successful bone regeneration. This article discusses various classes of biomaterials currently used in craniofacial reconstruction. Also reviewed are clinical applications of biomaterials as delivery agents for sustained release of stem cells, genes, and growth factors. Recent promising advancements in 3D printing and bioprinting techniques that seem to be promising for future clinical treatments for craniofacial reconstruction are covered. Relevant topics in the bone regeneration literature exemplifying the potential of biomaterials to repair bone defects are highlighted.
Collapse
Affiliation(s)
- Greeshma Thrivikraman
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, OHSU School of Dentistry, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Avathamsa Athirasala
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, OHSU School of Dentistry, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Chelsea Twohig
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, OHSU School of Dentistry, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Sunil Kumar Boda
- Mary and Dick Holland Regenerative Medicine Program, Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE 68198-5965, USA
| | - Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, OHSU School of Dentistry, 2730 SW Moody Avenue, Portland, OR 97201, USA; Department of Biomedical Engineering, OHSU School of Medicine, 3303 SW Bond Avenue, Portland, OR 97239, USA; OHSU Center for Regenerative Medicine, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
45
|
Sultana T, Amirian J, Park C, Lee SJ, Lee BT. Preparation and characterization of polycaprolactone–polyethylene glycol methyl ether and polycaprolactone–chitosan electrospun mats potential for vascular tissue engineering. J Biomater Appl 2017; 32:648-662. [DOI: 10.1177/0885328217733849] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Recently, natural polymers are frequently comingled with synthetic polymers either by physical or chemical modification to prepare numerous tissue-engineered graft with promising biological function, strength, and stability. The aim of this study was to determine the efficiency for vascular tissue engineering of two distinctly different mats, one that comprised polycaprolactone–polyethylene glycol methyl ether and other that comprised polycaprolactone–chitosan. Nano/microfibrous mats prepared from electro-spinning were characterized for fiber diameter, porosity, wettability, and mechanical strength. Biological efficacy on both biodegradable mats was assessed by rat bone marrow mesenchymal stem cells, and polycaprolactone–polyethylene glycol methyl ether showed feasibility for use as an inner layer by inducing endothelial-specific gene expression and polycaprolactone–chitosan as an outer layer on dual layered without sacrificing tensile strength, small-diameter blood vessels. Therefore, scaffolds fabricated from this research could be potential sources for tissue-engineered vascular graft and could also overcome the well-known drawbacks, such as thrombogenicity and stenosis, in managing vascular disease.
Collapse
Affiliation(s)
- Tamanna Sultana
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University 366-1, Ssangyoung-Dong, Cheonan City, ChungCheonNam-Do, Republic of Korea
| | - Jhaleh Amirian
- Institute of Tissue Regeneration, Soonchunhyang University 366-1, Ssangyoung-Dong, Cheonan City, ChungCheonNam-Do, Republic of Korea
| | - Chanmi Park
- Institute of Tissue Regeneration, Soonchunhyang University 366-1, Ssangyoung-Dong, Cheonan City, ChungCheonNam-Do, Republic of Korea
| | - Seung Jin Lee
- Department of Thoracic and Cardiovascular Surgery, Soonchunhyang University Hospital, 31, 6-gil, Cheonan City, ChungCheonNam-Do, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University 366-1, Ssangyoung-Dong, Cheonan City, ChungCheonNam-Do, Republic of Korea
- Institute of Tissue Regeneration, Soonchunhyang University 366-1, Ssangyoung-Dong, Cheonan City, ChungCheonNam-Do, Republic of Korea
| |
Collapse
|
46
|
Mondal S. Preparation, properties and applications of nanocellulosic materials. Carbohydr Polym 2017; 163:301-316. [DOI: 10.1016/j.carbpol.2016.12.050] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 12/17/2016] [Accepted: 12/20/2016] [Indexed: 10/20/2022]
|
47
|
Liu X, Zhang Y, Li S, Wang Y, Sun T, Li Z, Cai L, Wang X, Zhou L, Lai R. Study of a new bone-targeting titanium implant-bone interface. Int J Nanomedicine 2016; 11:6307-6324. [PMID: 27932879 PMCID: PMC5135286 DOI: 10.2147/ijn.s119520] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
New strategies involving bone-targeting titanium (Ti) implant-bone interface are required to enhance bone regeneration and osseointegration for orthopedic and dental implants, especially in osteoporotic subjects. In this study, a new dual-controlled, local, bone-targeting delivery system was successfully constructed by loading tetracycline-grafted simvastatin (SV)-loaded polymeric micelles in titania nanotube (TNT) arrays, and a bone-targeting Ti implant-bone interface was also successfully constructed by implanting the delivery system in vivo. The biological effects were evaluated both in vitro and in vivo. The results showed that Ti surfaces with TNT-bone-targeting micelles could promote cytoskeletal spreading, early adhesion, alkaline phosphatase activity, and extracellular osteocalcin concentrations of rat osteoblasts, with concomitant enhanced protein expression of bone morphogenetic protein (BMP)-2. A single-wall bone-defect implant model was established in normal and ovariectomized rats as postmenopausal osteoporosis models. Microcomputed tomography imaging and BMP-2 expression in vivo demonstrated that the implant with a TNT-targeting micelle surface was able to promote bone regeneration and osseointegration in both animal models. Therefore, beneficial biological effects were demonstrated both in vitro and in vivo, which indicated that the bone-targeting effects of micelles greatly enhance the bioavailability of SV on the implant-bone interface, and the provision of SV-loaded targeting micelles alone exhibits the potential for extensive application in improving local bone regeneration and osseointegration, especially in osteoporotic subjects.
Collapse
Affiliation(s)
- Xiangning Liu
- The Medical Center of Stomatology, The First Affiliated Hospital of Jinan University
| | - Ye Zhang
- The Medical Center of Stomatology, The First Affiliated Hospital of Jinan University
| | - Shaobing Li
- The Department of Oral Implantology, Guangdong Provincial Stomatological Hospital, Southern Medical University
| | - Yayu Wang
- Department of Cell Biology, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Ting Sun
- The Medical Center of Stomatology, The First Affiliated Hospital of Jinan University
| | - Zejian Li
- The Medical Center of Stomatology, The First Affiliated Hospital of Jinan University
| | - Lizhao Cai
- The Medical Center of Stomatology, The First Affiliated Hospital of Jinan University
| | - Xiaogang Wang
- Department of Cell Biology, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Lei Zhou
- The Department of Oral Implantology, Guangdong Provincial Stomatological Hospital, Southern Medical University
| | - Renfa Lai
- The Medical Center of Stomatology, The First Affiliated Hospital of Jinan University
| |
Collapse
|
48
|
Calcium Phosphates and Angiogenesis: Implications and Advances for Bone Regeneration. Trends Biotechnol 2016; 34:983-992. [PMID: 27481474 DOI: 10.1016/j.tibtech.2016.07.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 01/10/2023]
Abstract
Calcium phosphates (CaPs) are among the most utilized synthetic biomaterials for bone regeneration, largely owing to their established osteoconductive and osteoinductive properties. While angiogenesis is a crucial prerequisite to bone formation, research and applications for CaPs have not appreciated its crucial role. This review discusses how CaPs influence angiogenesis, and highlights promising strategies that address this topic. The objective is to draw attention to the gap in the literature and to highlight the importance of angiogenesis in CaP research, development, and use.
Collapse
|
49
|
Chen X, Liu Y, Miao L, Wang Y, Ren S, Yang X, Hu Y, Sun W. Controlled release of recombinant human cementum protein 1 from electrospun multiphasic scaffold for cementum regeneration. Int J Nanomedicine 2016; 11:3145-58. [PMID: 27471382 PMCID: PMC4948698 DOI: 10.2147/ijn.s104324] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Periodontitis is a major cause for tooth loss, which affects about 15% of the adult population. Cementum regeneration has been the crux of constructing the periodontal complex. Cementum protein 1 (CEMP1) is a cementum-specific protein that can induce cementogenic differentiation. In this study, poly(ethylene glycol) (PEG)-stabilized amorphous calcium phosphate (ACP) nanoparticles were prepared by wet-chemical method and then loaded with recombinant human CEMP1 (rhCEMP1) for controlled release. An electrospun multiphasic scaffold constituted of poly(ε-caprolactone) (PCL), type I collagen (COL), and rhCEMP1/ACP was fabricated. The effects of rhCEMP1/ACP/PCL/COL scaffold on the attachment proliferation, osteogenic, and cementogenic differentiations of human periodontal ligament cells, (PDLCs) were systematically investigated. A critical size defect rat model was introduced to evaluate the effect of tissue regeneration of the scaffolds in vivo. The results showed that PEG-stabilized ACP nanoparticles formed a core-shell structure with sustained release of rhCEMP1 for up to 4 weeks. rhCEMP1/ACP/PCL/COL scaffold could suppress PDLCs proliferation behavior and upregulate the expression of cementoblastic markers including CEMP1 and cementum attachment protein while downregulating osteoblastic markers including osteocalcin and osteopontin when it was cocultured with PDLCs in vitro for 7 days. Histology analysis of cementum after being implanted with the scaffold in rats for 8 weeks showed that there was cementum-like tissue formation but little bone formation. These results indicated the potential of using electrospun multiphasic scaffolds for controlled release of rhCEMP1 for promoting cementum regeneration in reconstruction of the periodontal complex.
Collapse
Affiliation(s)
- Xiaofeng Chen
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Yu Liu
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Leiying Miao
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Yangyang Wang
- Department of Materials Science and Engineering, College of Materials Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, People's Republic of China
| | - Shuangshuang Ren
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Xuebin Yang
- Biomaterials and Tissue Engineering Group, Leeds Dental Institute, University of Leeds, Leeds, UK
| | - Yong Hu
- Institute of Materials Engineering, National Laboratory of Solid State Micro Structure, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Weibin Sun
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
50
|
Tee BC, Desai KGH, Kennedy KS, Sonnichsen B, Kim DG, Fields HW, Mallery SR, Schwendeman SP, Sun Z. Reconstructing jaw defects with MSCs and PLGA-encapsulated growth factors. Am J Transl Res 2016; 8:2693-2704. [PMID: 27398152 PMCID: PMC4931163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/07/2016] [Indexed: 06/06/2023]
Abstract
Cell and growth factor-based tissue engineering has shown great potentials for skeletal regeneration. This study tested its feasibility in reconstructing large mandibular defects and compared the efficacy of varied construction materials and sealing methods. Bilateral mandibular critical-size (5-cm(3)) defects were created on six 4-month-old domestic pigs, and grafted with β-tricalcium phosphate (βTCP) only (Group-A), βTCP with autologous bone marrow-derived mesenchymal stem cells (BM-MSCs) (Group-B), and βTCP with BM-MSCs and biodegradable poly(lactic-co-glycolic acid) (PLGA) microspheres containing bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) (Group-C). The buccal sides of Groups-B/-C were either sealed by fibrin sealant or by a biodegradable PLGA barrier membrane before soft-tissue closure. Computed tomography (CT), microCT and histology analyses were performed 12 weeks postoperatively. In vitro data demonstrated that BM-MSCs, with MSC properties confirmed, remained vital after integration with βTCP; and PLGA microspheres exhibited an initial burst followed by slow and continuous release of growth factors over a period of 28 days. In vivo data demonstrated that Group-B/-C sites had significantly greater gap obliteration, higher tissue mineral densities and more residual βTCP granules (p<0.05, Kruskal-Wallis tests). Qualitatively, Group-B/-C defect sites had started remodeling while Group-A sites were mainly forming new bone to bridge the gaps. Furthermore, βTCP degradation was not mediated by macrophages or osteoclasts, and was significantly slowed down by sealing the defects with barrier membrane. Combined, these data present a promising formulation composed of βTCP granules, autologous MSCs, controlled-release growth factors and biodegradable PLGA barrier membrane for the reconstruction of critical-size mandibular defects.
Collapse
Affiliation(s)
- Boon Ching Tee
- Division of Biosciences, College of Dentistry, The Ohio State UniversityColumbus, Ohio, USA
| | - Kashappa Goud H Desai
- Department of Pharmaceutical Sciences and Biomedical Engineering, The Biointerfaces Institute, University of MichiganAnn Arbor, Michigan, USA
- Current Address: GlaxoSmithKlineKing of Prussia, PA 19406, USA
| | - Kelly S Kennedy
- Division of Oral and Maxillofacial Surgery, College of Dentistry, The Ohio State UniversityColumbus, Ohio, USA
| | - Brittany Sonnichsen
- Division of Oral and Maxillofacial Surgery, College of Dentistry, The Ohio State UniversityColumbus, Ohio, USA
| | - Do-Gyoon Kim
- Division of Orthodontics, College of Dentistry, The Ohio State UniversityColumbus, Ohio, USA
| | - Henry W Fields
- Division of Orthodontics, College of Dentistry, The Ohio State UniversityColumbus, Ohio, USA
| | - Susan R Mallery
- Division of Oral Pathology & Radiology, College of Dentistry, The Ohio State UniversityColumbus, Ohio, USA
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences and Biomedical Engineering, The Biointerfaces Institute, University of MichiganAnn Arbor, Michigan, USA
| | - Zongyang Sun
- Division of Orthodontics, College of Dentistry, The Ohio State UniversityColumbus, Ohio, USA
| |
Collapse
|