401
|
Liang W, Han B, Hai Y, Sun D, Yin P. Mechanism of Action of Mesenchymal Stem Cell-Derived Exosomes in the Intervertebral Disc Degeneration Treatment and Bone Repair and Regeneration. Front Cell Dev Biol 2022; 9:833840. [PMID: 35096846 PMCID: PMC8795890 DOI: 10.3389/fcell.2021.833840] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Exosomes are extracellular vesicles formed by various donor cells that regulate gene expression and cellular function in recipient cells. Exosomes derived from mesenchymal stem cells (MSC-Exos) perform the regulatory function of stem cells by transporting proteins, nucleic acids, and lipids. Intervertebral disc degeneration (IDD) is one of the main causes of low back pain, and it is characterized by a decreased number of nucleus pulposus cells, extracellular matrix decomposition, aging of the annulus fibrosus, and cartilage endplate calcification. Besides, nutrient transport and structural repair of intervertebral discs depend on bone and cartilage and are closely related to the state of the bone. Trauma, disease and aging can all cause bone injury. However, there is a lack of effective drugs against IDD and bone injury. Recent MSC-Exos fine tuning has led to significant progress in the IDD treatment and bone repair and regeneration. In this review, we looked at the uniqueness of MSC-Exos, and the potential treatment mechanisms of MSC-Exos with respect to IDD, bone defects and injuries.
Collapse
Affiliation(s)
- Weishi Liang
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Bo Han
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yong Hai
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Duan Sun
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Peng Yin
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
402
|
Naredla M, Osmani RA, S M, Gupta MS, Gowda DV. Potential applications of coral sand in bone healing and drug delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
403
|
Abazari MF, Torabinejad S, Karizi SZ, Samadian H, Jalili-ghelichi S, Norouzi M, Askari H, Poortahmasebi V, Enderami SE, Soleimanifar F. Biologically modified electrospun polycaprolactone nanofibrous scaffold promotes osteogenic differentiation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.103050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
404
|
Huber J, Griffin MF, Longaker MT, Quarto N. Exosomes: A Tool for Bone Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:101-113. [PMID: 33297857 PMCID: PMC8892957 DOI: 10.1089/ten.teb.2020.0246] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Mesenchymal stem cells (MSCs) have been repeatedly shown to be a valuable source for cell-based therapy in regenerative medicine, including bony tissue repair. However, engraftment at the injury site is poor. Recently, it has been suggested that MSCs and other cells act through a paracrine signaling mechanism. Exosomes are nanostructures that have been implicated in this process. They carry DNA, RNA, proteins, and lipids and play an important role in cell-to-cell communication directly modulating their target cell at a transcriptional level. In a bone microenvironment, they have been shown to increase osteogenesis and osteogenic differentiation in vivo and in vitro. In the following review, we will discuss the most advanced and significant knowledge of biological functions of exosomes in bone regeneration and their clinical applications in osseous diseases. Impact statement Mesenchymal stem cells have been shown to be a promising tool in bone tissue engineering. Recently, it has been suggested that they secrete exosomes containing messenger RNA, proteins, and lipids, thus acting through paracrine signaling mechanisms. Considering that exosomes are nonteratogenic and have low immunogenic potential, they could potentially replace stem-cell based therapy and thus eradicate the risk of neoplastic transformation associated with cell transplantations in bone regeneration.
Collapse
Affiliation(s)
- Julika Huber
- Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA.,Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany.,Address correspondence to: Julika Huber, MD, Dr. med, Hagey Laboratory for Pediatric Regenerative Medicine, School of Medicine, Stanford University, 257 Campus Drive, Stanford, CA 94305-5148, USA
| | - Michelle F. Griffin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA.,Stanford Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Natalina Quarto
- Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA.,Dipartimento di Scienze Biomediche Avanzate, Universita’ degli Studi di Napoli Federico II, Napoli, Italy.,Address correspondence to: Natalina Quarto, PhD, Hagey Laboratory for Pediatric Regenerative Medicine, School of Medicine, Stanford University, 257 Campus Drive, Stanford, CA 94305-5148, USA
| |
Collapse
|
405
|
Petre DG, Leeuwenburgh SCG. The Use of Fibers in Bone Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:141-159. [PMID: 33375900 DOI: 10.1089/ten.teb.2020.0252] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Bone tissue engineering aims to restore and maintain the function of bone by means of biomaterial-based scaffolds. This review specifically focuses on the use of fibers in biomaterials used for bone tissue engineering as suitable environment for bone tissue repair and regeneration. We present a bioinspired rationale behind the use of fibers in bone tissue engineering and provide an overview of the most common fiber fabrication methods, including solution, melt, and microfluidic spinning. Subsequently, we provide a brief overview of the composition of fibers that are used in bone tissue engineering, including fibers composed of (i) natural polymers (e.g., cellulose, collagen, gelatin, alginate, chitosan, and silk, (ii) synthetic polymers (e.g., polylactic acid [PLA], polycaprolactone, polyglycolic acid [PGA], polyethylene glycol, and polymer blends of PLA and PGA), (iii) ceramic fibers (e.g., aluminium oxide, titanium oxide, and zinc oxide), (iv) metallic fibers (e.g., titanium and its alloys, copper and magnesium), and (v) composite fibers. In addition, we review the most relevant fiber modification strategies that are used to enhance the (bio)functionality of these fibers. Finally, we provide an overview of the applicability of fibers in biomaterials for bone tissue engineering, with a specific focus on mechanical, pharmaceutical, and biological properties of fiber-functionalized biomaterials for bone tissue engineering. Impact statement Natural bone is a complex composite material composed of an extracellular matrix of mineralized fibers containing living cells and bioactive molecules. Consequently, the use of fibers in biomaterial-based scaffolds offers a wide variety of opportunities to replicate the functional performance of bone. This review provides an overview of the use of fibers in biomaterials for bone tissue engineering, thereby contributing to the design of novel fiber-functionalized bone-substituting biomaterials of improved functionality regarding their mechanical, pharmaceutical, and biological properties.
Collapse
Affiliation(s)
- Daniela Geta Petre
- Department of Dentistry-Regenerative Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Sander C G Leeuwenburgh
- Department of Dentistry-Regenerative Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
406
|
Juhl OJ, Merife A, Zhang Y, Donahue HJ. Inhibition of focal adhesion turnover prevents osteoblastic differentiation through β‐catenin mediated transduction of pro‐osteogenic substrate. J Biomed Mater Res B Appl Biomater 2022; 110:1573-1586. [PMID: 35099117 PMCID: PMC9306686 DOI: 10.1002/jbm.b.35018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 11/24/2022]
Abstract
The mechanism by which substrate surface characteristics are transduced by osteoblastic cells and their progenitors is not fully known. Data from previous studies by our group suggest the involvement of β‐catenin in the mechanism by which substrate surface characteristics are transduced. This focal adhesion and β‐catenin mediated mechanism functions through the liberation of β‐catenin from focal adhesion complexes in response to pro‐osteogenic substrate (POS) characteristics. After liberation, β‐catenin translocates and facilitates upregulation of genes associated with osteogenesis. It is not known whether the observed correlation between focal adhesion turnover and β‐catenin translocation directly results from focal adhesion turnover. In this study we inhibited focal adhesion turnover using a focal adhesion kinase inhibitor PF‐573228. We found that inhibition of focal adhesion turnover resulted in an abrogation of the more rapid translocation and increased transcriptional activity of β‐catenin induced by POS. In addition, inhibition of focal adhesion turnover mitigated the increase in osteoblastic differentiation induced by a POS as measured by alkaline phosphatase enzymatic activity and osteogenic gene and protein expression. Together, these data, coupled with previous findings, suggest that the observed β‐catenin translocation is a result of focal adhesion turnover, providing evidence for a focal adhesion initiated, β‐catenin mediated mechanism of substrate surface signal transduction.
Collapse
Affiliation(s)
- Otto J. Juhl
- Department of Biomedical Engineering and Institute for Engineering and Medicine Virginia Commonwealth University Richmond Virginia USA
| | - Anna‐Blessing Merife
- Department of Biomedical Engineering and Institute for Engineering and Medicine Virginia Commonwealth University Richmond Virginia USA
| | - Yue Zhang
- Department of Biomedical Engineering and Institute for Engineering and Medicine Virginia Commonwealth University Richmond Virginia USA
| | - Henry J. Donahue
- Department of Biomedical Engineering and Institute for Engineering and Medicine Virginia Commonwealth University Richmond Virginia USA
| |
Collapse
|
407
|
Reyna-Urrutia VA, González-González AM, Rosales-Ibáñez R. Compositions and Structural Geometries of Scaffolds Used in the Regeneration of Cleft Palates: A Review of the Literature. Polymers (Basel) 2022; 14:polym14030547. [PMID: 35160534 PMCID: PMC8840587 DOI: 10.3390/polym14030547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Cleft palate (CP) is one of the most common birth defects, presenting a multitude of negative impacts on the health of the patient. It also leads to increased mortality at all stages of life, economic costs and psychosocial effects. The embryological development of CP has been outlined thanks to the advances made in recent years due to biomolecular successions. The etiology is broad and combines certain environmental and genetic factors. Currently, all surgical interventions work off the principle of restoring the area of the fissure and aesthetics of the patient, making use of bone substitutes. These can involve biological products, such as a demineralized bone matrix, as well as natural–synthetic polymers, and can be supplemented with nutrients or growth factors. For this reason, the following review analyzes different biomaterials in which nutrients or biomolecules have been added to improve the bioactive properties of the tissue construct to regenerate new bone, taking into account the greatest limitations of this approach, which are its use for bone substitutes for large areas exclusively and the lack of vascularity. Bone tissue engineering is a promising field, since it favors the development of porous synthetic substitutes with the ability to promote rapid and extensive vascularization within their structures for the regeneration of the CP area.
Collapse
|
408
|
Becerra J, Rodriguez M, Leal D, Noris-Suarez K, Gonzalez G. Chitosan-collagen-hydroxyapatite membranes for tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:18. [PMID: 35072812 PMCID: PMC8786760 DOI: 10.1007/s10856-022-06643-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/05/2022] [Indexed: 05/17/2023]
Abstract
Tissue engineering is growing in developing new technologies focused on providing effective solutions to degenerative pathologies that affect different types of connective tissues. The search for biocompatible, bioactive, biodegradable, and multifunctional materials has grown significantly in recent years. Chitosan, calcium phosphates collagen, and their combination as composite materials fulfill the required properties and could result in biostimulation for tissue regeneration. In the present work, the chitosan/collagen/hydroxyapatite membranes were prepared with different concentrations of collagen and hydroxyapatite. Cell adhesion was evaluated by MTS assay for two in vitro models. Additionally, cytotoxicity of the different membranes employing hemolysis of erythrocytes isolated from human blood was carried out. The structure of the membranes was analyzed by X-rays diffraction (XRD) and Fourier transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), and thermal stability properties by thermogravimetric methods (TGA). The highest cell adhesion after 48 h was obtained for chitosan membranes with the highest hydroxyapatite and collagen content. All composite membranes showed good cell adhesion and low cytotoxicity, suggesting that these materials have a significant potential to be used as biomaterials for tissue engineering. Graphical abstract.
Collapse
Affiliation(s)
- José Becerra
- Instituto de Ciencias Básicas, Universidad Técnica de Manabí, Portoviejo, Ecuador
- Lab. de Materiales, Centro de Ingeniería de Materiales y Nanotecnología, Instituto Venezolano de Investigaciones Científicas, IVIC, Caracas, Venezuela
| | | | - Dayana Leal
- Instituto de Ciencias Básicas, Universidad Técnica de Manabí, Portoviejo, Ecuador
| | | | - Gema Gonzalez
- Lab. de Materiales, Centro de Ingeniería de Materiales y Nanotecnología, Instituto Venezolano de Investigaciones Científicas, IVIC, Caracas, Venezuela.
- Yachay Tech University, School of Physical Sciences and Nanotechnology, Urcuqui, 100119, Ecuador.
| |
Collapse
|
409
|
Gao Q, Wang L, Wang S, Huang B, Jing Y, Su J. Bone Marrow Mesenchymal Stromal Cells: Identification, Classification, and Differentiation. Front Cell Dev Biol 2022; 9:787118. [PMID: 35047499 PMCID: PMC8762234 DOI: 10.3389/fcell.2021.787118] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/25/2021] [Indexed: 12/20/2022] Open
Abstract
Bone marrow mesenchymal stromal cells (BMSCs), identified as pericytes comprising the hematopoietic niche, are a group of heterogeneous cells composed of multipotent stem cells, including osteochondral and adipocyte progenitors. Nevertheless, the identification and classification are still controversial, which limits their application. In recent years, by lineage tracing and single-cell sequencing, several new subgroups of BMSCs and their roles in normal physiological and pathological conditions have been clarified. Key regulators and mechanisms controlling the fate of BMSCs are being revealed. Cross-talk among subgroups of bone marrow mesenchymal cells has been demonstrated. In this review, we focus on recent advances in the identification and classification of BMSCs, which provides important implications for clinical applications.
Collapse
Affiliation(s)
- Qianmin Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China.,Shanghai University Institute of Advanced Interdisciplinary Materials Science, Shanghai, China
| | - Lipeng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Biaotong Huang
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,Shanghai University Institute of Advanced Interdisciplinary Materials Science, Shanghai, China.,Wenzhou Institute of Shanghai University, Wenzhou, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,Shanghai University Institute of Advanced Interdisciplinary Materials Science, Shanghai, China
| | - Jiacan Su
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
410
|
Kareem MM, Tanner KE. Methods of producing three dimensional electrospun scaffolds for bone tissue engineering: A review. Proc Inst Mech Eng H 2022; 236:9544119211069463. [PMID: 35048771 DOI: 10.1177/09544119211069463] [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] [Indexed: 02/21/2024]
Abstract
Bone is a dynamic, living tissue that exists and renews itself continuously in a 3D manner. Nevertheless, complex clinical conditions require a bone substitute to replace the defective bone and/or accelerate bone healing. Bone tissue engineering aims to treat bone defects that fail to heal on their own. Electrospinning provides an opportunity to create nano- to micro-fibrous scaffolds that mimic the architecture of the natural extracellular matrix (ECM) with high porosity and large specific surface area. Despite these advantages, traditional electrospun meshes can only provide a 2D architecture for cell attachment and proliferation rather than the 3D attachment in native tissue. Fabrication of 3D electrospun scaffolds for bone tissue regeneration is a challenging task, which has attracted significant attention over the past couple of decades. This review highlights recent strategies used to produce 3D electrospun/co-electrospun scaffolds for bone tissue applications describing the materials and procedures. It also considers combining conventional and coaxial electrospinning with other scaffold manufacturing techniques to produce 3D structures which have the potential to engineer missing bone in the human body.Graphical abstract[Formula: see text].
Collapse
Affiliation(s)
- Muna M Kareem
- Department of Medical Instrumentation Techniques Engineering, Dijlah University College, Baghdad, Iraq
| | - K E Tanner
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| |
Collapse
|
411
|
Fosca M, Rau JV, Uskoković V. Factors influencing the drug release from calcium phosphate cements. Bioact Mater 2022; 7:341-363. [PMID: 34466737 PMCID: PMC8379446 DOI: 10.1016/j.bioactmat.2021.05.032] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/19/2022] Open
Abstract
Thanks to their biocompatibility, biodegradability, injectability and self-setting properties, calcium phosphate cements (CPCs) have been the most economical and effective biomaterials of choice for use as bone void fillers. They have also been extensively used as drug delivery carriers owing to their ability to provide for a steady release of various organic molecules aiding the regeneration of defective bone, including primarily antibiotics and growth factors. This review provides a systematic compilation of studies that reported on the controlled release of drugs from CPCs in the last 25 years. The chemical, compositional and microstructural characteristics of these systems through which the control of the release rates and mechanisms could be achieved have been discussed. In doing so, the effects of (i) the chemistry of the matrix, (ii) porosity, (iii) additives, (iv) drug types, (v) drug concentrations, (vi) drug loading methods and (vii) release media have been distinguished and discussed individually. Kinetic specificities of in vivo release of drugs from CPCs have been reviewed, too. Understanding the kinetic and mechanistic correlations between the CPC properties and the drug release is a prerequisite for the design of bone void fillers with drug release profiles precisely tailored to the application area and the clinical picture. The goal of this review has been to shed light on these fundamental correlations.
Collapse
Affiliation(s)
- Marco Fosca
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere 100, 00133, Rome, Italy
| | - Julietta V. Rau
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere 100, 00133, Rome, Italy
- I.M. Sechenov First Moscow State Medical University, Institute of Pharmacy, Department of Analytical, Physical and Colloid Chemistry, Trubetskaya 8, build. 2, 119991, Moscow, Russia
| | - Vuk Uskoković
- Advanced Materials and Nanobiotechnology Laboratory, TardigradeNano LLC, Irvine, CA 92604, United States
| |
Collapse
|
412
|
Wang S, Gu R, Wang F, Zhao X, Yang F, Xu Y, Yan F, Zhu Y, Xia D, Liu Y. 3D-Printed PCL/Zn scaffolds for bone regeneration with a dose-dependent effect on osteogenesis and osteoclastogenesis. Mater Today Bio 2022; 13:100202. [PMID: 35036897 PMCID: PMC8753274 DOI: 10.1016/j.mtbio.2021.100202] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 12/16/2022] Open
Abstract
Polycaprolactone (PCL) is a polymer material suitable for being prepared into porous scaffolds used in bone tissue engineering, however, insufficient osteogenic ability and mechanical strength limit its application. Zinc (Zn) alloy with proper mechanical strength and osteogenesis is a promising biodegradable metal that have attracted much attention. Herein, we combined the advantages of PCL and Zn by fabricating PCL/Zn composite scaffolds with different Zn powder contents (1 wt%, 2 wt%, 3 wt%) through fused deposition modelling. The mechanical property, cytocompatibility and Zn ions release behavior of PCL/Zn scaffolds were analyzed in vitro. The osteogenesis and osteoclastogenesis properties of the scaffolds were evaluated by being implanted into Sprague-Dawley rats calvaria defect. Results showed that the PCL/Zn scaffolds exhibited improved mechanical properties and cytocompatibility compared with the pure PCL scaffolds. At 8 weeks after in vivo implantaion, the addition of Zn powder promoted new bone formation, in a dose-dependent manner. The scaffolds with 2 wt% Zn displayed the best osteogenic effect, while the osteogenic effect was slightly reduced in the scaffolds with 3 wt% Zn. In the studied Zn contents, the PCL/Zn scaffolds gradually promoted osteoclastogenesis with increasd Zn content. In the 3 wt% Zn group, TRAP-positive cells were observed on the newly formed bone edges around the scaffolds. These dose-dependent effects were verified in vitro using MC3T3-E1 and RAW264.7 cells. Finally, we revealed that Zn2+ regulated osteogenesis and osteoclastogenesis by activation of the Wnt/β-catenin and NF-κB signalling pathways, respectively.
Collapse
Affiliation(s)
- Siyi Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Ranli Gu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Feilong Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Xiao Zhao
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Fan Yang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Yuqian Xu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Fanyu Yan
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Dandan Xia
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Department of Dental Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| |
Collapse
|
413
|
Ladeira B, Custodio C, Mano J. Core-Shell Microcapsules: Biofabrication and Potential Applications in Tissue Engineering and Regenerative Medicine. Biomater Sci 2022; 10:2122-2153. [DOI: 10.1039/d1bm01974k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The construction of biomaterial scaffolds that accurately recreate the architecture of living tissues in vitro is a major challenge in the field of tissue engineering and regenerative medicine. Core-shell microcapsules...
Collapse
|
414
|
Depboylu FN, Korkusuz P, Yasa E, Korkusuz F. Smart Bioceramics for Orthopedic Applications. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2022:157-186. [DOI: 10.1007/978-981-16-7439-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
415
|
Meng Y, Chen J, Wu X. Effect of Hypoxia-Inducible Factor-1 α on Osteogenesis of Titanium Dioxide Nanotube Bone Marrow Mesenchymal Stem Cells with Different Diameters Under Periodic Tensile Stress. J Biomed Nanotechnol 2022; 18:112-119. [PMID: 35180904 DOI: 10.1166/jbn.2022.3221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Bone marrow mesenchymal stem cells (BMSC) have the ability to multi polarize with multiple tropisms and participate in tissue remodeling. This study assessed the effect of titanium dioxide nanotubes with different diameters on ossification of BMSC cells and HIF-1α expression in BMSC ossification. Titanium dioxide nanotubes with different diameters were prepared and then the following groups were set according to the size of pressure; Ti group, NT10 group, NT30 group, and NT60 group. Analysis of cell morphology was done by fluorescence microscope, while adhesion and proliferation were assessed by MTT assay. Moreover, ALP activity, collagen secretion and outer matrix mineralization and expression of HIF-1α, VEGF, and TWIST were assessed by RT-PCR and Western blot. The P3 generation of BMSC cells was successfully obtained. Three types of nanotubes were arranged regularly and contact angle showed NT60<NT30<NT10<Ti, protein adsorption presented NT10>Ti>NT30>NT60 (P < 0.05). Cells from NT30 and N60 groups showed obvious expansion with pseudopodia and pseudo plates of cells. Cell adhesion showed changes in sizes of NT10>Ti>NT30>NT66. NT60 group showed lower cell proliferation and higher ALP activity and collagen secretion than the other groups. NT30 and NT60 group presented higher mineralization level, larger diameter, and higher degree of promotion. The NT30 group presented lowest content of HIF-1α (0.12 ± 0.03), VEGF (0.013 ± 0.004), and TWIST (0.014 ± 0.003). Inoculation of BMSCs on titanium dioxide nanotubes of different diameters under cyclical tensile stress environment can promote growth of BMSC cells in a diameter-dependent manner.
Collapse
Affiliation(s)
- Yu Meng
- Department of Emergency Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Juwu Chen
- Department of Emergency Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xuejian Wu
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan, China
| |
Collapse
|
416
|
Shape optimization of orthopedic porous scaffolds to enhance mechanical performance. J Mech Behav Biomed Mater 2022; 128:105098. [DOI: 10.1016/j.jmbbm.2022.105098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/02/2021] [Accepted: 01/17/2022] [Indexed: 11/19/2022]
|
417
|
Li S, Huan Y, Zhu B, Chen H, Tang M, Yan Y, Wang C, Ouyang Z, Li X, Xue J, Wang W. Research progress on the biological modifications of implant materials in 3D printed intervertebral fusion cages. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 33:2. [PMID: 34940930 PMCID: PMC8702412 DOI: 10.1007/s10856-021-06609-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 10/06/2021] [Indexed: 05/26/2023]
Abstract
Anterior spine decompression and reconstruction with bone grafts and fusion is a routine spinal surgery. The intervertebral fusion cage can maintain intervertebral height and provide a bone graft window. Titanium fusion cages are the most widely used metal material in spinal clinical applications. However, there is a certain incidence of complications in clinical follow-ups, such as pseudoarticulation formation and implant displacement due to nonfusion of bone grafts in the cage. With the deepening research on metal materials, the properties of these materials have been developed from being biologically inert to having biological activity and biological functionalization, promoting adhesion, cell differentiation, and bone fusion. In addition, 3D printing, thin-film, active biological material, and 4D bioprinting technology are also being used in the biofunctionalization and intelligent advanced manufacturing processes of implant devices in the spine. This review focuses on the biofunctionalization of implant materials in 3D printed intervertebral fusion cages. The surface modifications of implant materials in metal endoscopy, material biocompatibility, and bioactive functionalizationare summarized. Furthermore, the prospects and challenges of the biofunctionalization of implant materials in spinal surgery are discussed. Fig.a.b.c.d.e.f.g As a pre-selected image for the cover, I really look forward to being selected. Special thanks to you for your comments.
Collapse
Affiliation(s)
- Shan Li
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
- Plastic and Cosmetic Surgery, Hunan Want Want Hospital, Changsha, China
| | - Yifan Huan
- R&D Department, Hunan Yuanpin Cell Biotechnology Co. Ltd., Changsha, China
| | - Bin Zhu
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Haoxiang Chen
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Ming Tang
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Yiguo Yan
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Cheng Wang
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Zhihua Ouyang
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Xuelin Li
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Jingbo Xue
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China.
| | - Wenjun Wang
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China.
| |
Collapse
|
418
|
Dixon DT, Gomillion CT. Conductive Scaffolds for Bone Tissue Engineering: Current State and Future Outlook. J Funct Biomater 2021; 13:1. [PMID: 35076518 PMCID: PMC8788550 DOI: 10.3390/jfb13010001] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022] Open
Abstract
Bone tissue engineering strategies attempt to regenerate bone tissue lost due to injury or disease. Three-dimensional (3D) scaffolds maintain structural integrity and provide support, while improving tissue regeneration through amplified cellular responses between implanted materials and native tissues. Through this, scaffolds that show great osteoinductive abilities as well as desirable mechanical properties have been studied. Recently, scaffolding for engineered bone-like tissues have evolved with the use of conductive materials for increased scaffold bioactivity. These materials make use of several characteristics that have been shown to be useful in tissue engineering applications and combine them in the hope of improved cellular responses through stimulation (i.e., mechanical or electrical). With the addition of conductive materials, these bioactive synthetic bone substitutes could result in improved regeneration outcomes by reducing current factors limiting the effectiveness of existing scaffolding materials. This review seeks to overview the challenges associated with the current state of bone tissue engineering, the need to produce new grafting substitutes, and the promising future that conductive materials present towards alleviating the issues associated with bone repair and regeneration.
Collapse
Affiliation(s)
- Damion T. Dixon
- School of Environmental, Civil, Agricultural and Mechanical Engineering, University of Georgia, Athens, GA 30602, USA;
| | - Cheryl T. Gomillion
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
419
|
Polymer-Based Bone Substitutes in Periodontal Infrabony Defects: A Systematic Evaluation of Clinical Studies. Polymers (Basel) 2021; 13:polym13244445. [PMID: 34960996 PMCID: PMC8705724 DOI: 10.3390/polym13244445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/07/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022] Open
Abstract
Background and Objectives: The aim was to systematically review the available literature regarding the use of polymers as a bone substitute for the treatment of periodontal infrabony defect. Materials and methods: Three databases (PubMed, Scopus and Web of Science) were searched to find all relevant studies published in English from inception until September 2021 using a combination of keywords. The inclusion criteria consisted of human clinical studies which reported the use of a polymer-based bone substitute in the treatment of infrabony defects. Results: 164 studies were provided from the databases. Of these, five articles were eligible and reported favorable outcome in terms of probing depth, clinical attachment gain and defect fill at the follow-up (3 months and 6 months). Conclusions: Polymer based-bone substitutes may represent a useful alternative in treating infrabony defects. Due to the limited number of studies, more research is needed to sustain the advantages of these products.
Collapse
|
420
|
Biofunctional phosphorylated magnetic scaffold for bone tissue engineering. Colloids Surf B Biointerfaces 2021; 211:112284. [PMID: 34952284 DOI: 10.1016/j.colsurfb.2021.112284] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/23/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022]
Abstract
The development of highly bioactive engineered scaffolds is required to promote bone regeneration and the success of bone tissue engineering treatment approaches. This study attempts to fabricate a biofunctional magnetic scaffold based on new phosphorylated polycaprolactone combined with gelatin (MNPs-PCL-P/gelatin). Phosphorylated polymer and magnetic nanoparticles (MNPs) were synthesized and characterized by NMR, FT-IR, TEM, and DLS instruments. The synthetic polymer, MNPs, and biopolymer were mixed then freeze-dried to prepare a porous scaffold. Physiochemical assessments showed that a scaffold with well-developed porous morphology, and stable structure was obtained. MNPs-PCL-P/gelatin scaffold had no toxicity on human dental pulp stem cells (hDPSCs). The use of phosphorous-containing polymer resulted in improvement of the scaffold's osteoconductivity to support proper cell attachment and promote cell proliferation. Phosphate group by mimicking function of bone phosphate groups stimulate bone mineralization that reflected by alizarin red S staining assay. The presence of MNPs resulted in higher ALP activity and increased expression level of RUNX2, BMP2 osteogenic biomarkers. Also, phosphorylation enhanced osteoinductivity of scaffold and upregulate RUNX2, BMP2, COL1A1, and OCN genes in phosphors-containing scaffold test groups. It seems that biocompatible MNPs-PCL-P/gelatin scaffold possesses the potential of applications in bone tissue engineering.
Collapse
|
421
|
Evaluation of Stress Distribution during Insertion of Tapered Dental Implants in Various Osteotomy Techniques: Three-Dimensional Finite Element Study. MATERIALS 2021; 14:ma14247547. [PMID: 34947142 PMCID: PMC8704667 DOI: 10.3390/ma14247547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 11/17/2022]
Abstract
Conventional osteotomy techniques can, in some cases, induce higher stress on bone during implant insertion as a result of higher torque. The aim of the present study was to evaluate and compare the stress exerted on the underlying osseous tissues during the insertion of a tapered implant using different osteotomy techniques through a dynamic finite element analysis which has been widely applied to study biomedical problems through computer-aided software. In three different types of osteotomy techniques, namely conventional (B1), bone tap (B2), and countersink (B3), five models and implants designed per technique were prepared, implant insertion was simulated, and stress exerted by the implant during each was evaluated. Comparison of stress scores on the cortical and cancellous bone at different time points and time intervals from initiation of insertion to the final placement of the implant was made. There was a highly statistically significant difference between B1 and B2 (p = 0.0001) and B2 and B3 (p = 0.0001) groups. In contrast, there was no statistically significant difference in the stress scores between B1 and B3 (p = 0.3080) groups at all time points of implant placement. Overall, a highly significant difference was observed between the stresses exerted in each technique. Within the limitations of our study, bone tap significantly exerted lesser stresses on the entire bone than conventional and countersink type of osteotomy procedures. Considering the stress distribution at the crestal region, the countersink showed lower values in comparison to others.
Collapse
|
422
|
Sidharthan DS, Abhinandan R, Balagangadharan K, Selvamurugan N. Advancements in nucleic acids-based techniques for bone regeneration. Biotechnol J 2021; 17:e2100570. [PMID: 34882984 DOI: 10.1002/biot.202100570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 12/21/2022]
Abstract
The dynamic biology of bone involving an enormous magnitude of cellular interactions and signaling transduction provides ample biomolecular targets, which can be enhanced or repressed to mediate a rapid regeneration of the impaired bone tissue. The delivery of nucleic acids such as DNA and RNA can enhance the expression of osteogenic proteins. Members of the RNA interference pathway such as miRNA and siRNA can repress negative osteoblast differentiation regulators. Advances in nanomaterials have provided researchers with a plethora of delivery modules that can ensure proper transfection. Combining the nucleic acid carrying vectors with bone scaffolds has met with tremendous success in accomplishing bone formation. Recent years have witnessed the advent of CRISPR and DNA nanostructures in regenerative medicine. This review focuses on the delivery of nucleic acids and touches upon the prospect of CRISPR and DNA nanostructures for bone tissue engineering, emphasizing their potential in treating bone defects.
Collapse
Affiliation(s)
- Dharmaraj Saleth Sidharthan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ranganathan Abhinandan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Kalimuthu Balagangadharan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
423
|
Pańtak P, Cichoń E, Czechowska J, Zima A. Influence of Natural Polysaccharides on Properties of the Biomicroconcrete-Type Bioceramics. MATERIALS (BASEL, SWITZERLAND) 2021; 14:7496. [PMID: 34947091 PMCID: PMC8708244 DOI: 10.3390/ma14247496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/23/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022]
Abstract
In this paper, novel hybrid biomicroconcrete-type composites were developed and investigated. The solid phase of materials consisted of a highly reactive α -tricalcium phosphate (α-TCP) powder, hybrid hydroxyapatite-chitosan (HAp-CTS) material in the form of powder and granules (as aggregates), and the polysaccharides sodium alginate (SA) or hydroxypropyl methylcellulose (HPMC). The liquid/gel phase in the studied materials constituted a citrus pectin gel. The influence of SA or HPMC on the setting reaction, microstructure, mechanical as well as biological properties of biomicroconcretes was investigated. Studies revealed that manufactured cement pastes were characterized by high plasticity and cohesion. The dual setting system of developed biomicroconcretes, achieved through α-TCP setting reaction and polymer crosslinking, resulted in a higher compressive strength. Material with the highest content of sodium alginate possessed the highest mechanical strength (~17 MPa), whereas the addition of hydroxypropyl methylcellulose led to a subtle compressive strength decrease. The obtained biomicroconcretes were chemically stable and characterized by a high bioactive potential. The novel biomaterials with favorable physicochemical and biological properties can be prosperous materials for filling bone tissue defects of any shape and size.
Collapse
Affiliation(s)
| | | | | | - Aneta Zima
- Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza Av. 30, 30-058 Kraków, Poland; (P.P.); (E.C.); (J.C.)
| |
Collapse
|
424
|
McIvor MJ, Sharma PK, Birt CE, McDowell H, Wilson S, McKillop S, Acheson JG, Boyd AR, Meenan BJ. Direct monitoring of single-cell response to biomaterials by Raman spectroscopy. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:148. [PMID: 34862915 PMCID: PMC8643295 DOI: 10.1007/s10856-021-06624-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 11/03/2021] [Indexed: 06/13/2023]
Abstract
There is continued focus on the development of new biomaterials and associated biological testing methods needed to reduce the time taken for their entry to clinical use. The application of Raman spectroscopy to the study of individual cells that have been in contact with biomaterials offers enhanced in vitro information in a potentially non-destructive testing regime. The work presented here reports the Raman spectral analysis of discreet U-2 OS bone cells after exposure to hydroxyapatite (HA) coated titanium (Ti) substrates in both the as-deposited and thermally annealed states. These data show that cells that were in contact with the bioactive HA surface for 7 days had spectral markers similar to those cultured on the Ti substrate control for the same period. However, the spectral features for those cells that were in contact with the annealed HA surface had indicators of significant differentiation at day 21 while cells on the as-deposited surface did not show these Raman changes until day 28. The cells adhered to pristine Ti control surface showed no spectral changes at any of the timepoints studied. The validity of these spectroscopic results has been confirmed using data from standard in vitro cell viability, adhesion, and proliferation assays over the same 28-day culture period. In this case, cell maturation was evidenced by the formation of natural bone apatite, which precipitated intracellularly for cells exposed to both types of HA-coated Ti at 21 and 28 days, respectively. The properties of the intracellular apatite were markedly different from that of the synthetic HA used to coat the Ti substrate with an average particle size of 230 nm, a crystalline-like shape and Ca/P ratio of 1.63 ± 0.5 as determined by SEM-EDX analysis. By comparison, the synthetic HA particles used as a control had an average size of 372 nm and were more-rounded in shape with a Ca/P ratio of 0.8 by XPS analysis and 1.28 by SEM-EDX analysis. This study shows that Raman spectroscopy can be employed to monitor single U-2 OS cell response to biomaterials that promote cell maturation towards de novo bone thereby offering a label-free in vitro testing method that allows for non-destructive analyses.
Collapse
Affiliation(s)
- Mary Josephine McIvor
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK.
| | - Preetam K Sharma
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
- Department of Chemical Engineering, Loughborough University, Loughborough, LE11 3TU, England, UK
| | - Catherine E Birt
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| | - Hayley McDowell
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| | - Shannon Wilson
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| | - Stephen McKillop
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| | - Jonathan G Acheson
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| | - Adrian R Boyd
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| | - Brian J Meenan
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| |
Collapse
|
425
|
Veiga A, Castro F, Rocha F, Oliveira AL. An update on hydroxyapatite/collagen composites: What is there left to say about these bioinspired materials? J Biomed Mater Res B Appl Biomater 2021; 110:1192-1205. [PMID: 34860461 DOI: 10.1002/jbm.b.34976] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/25/2021] [Accepted: 11/16/2021] [Indexed: 01/01/2023]
Abstract
Hydroxyapatite (HAp)/collagen-based composite materials have been a constant in the development of bioinspired materials for bone tissue engineering. The most fundamental research works focus on combining HAp, due to its chemical similarity with the mineral component of bones, and collagen, which is the most abundant protein in the body. Modern studies have explored different two-dimensional (2D) and 3D structures, in order to obtain biomaterials with specific physicochemical, mechanical, and biological characteristics that can be applied in distinct biomedical applications. However, as there is already so much work developed with these materials, it is crucial to question: what can still be done? What is the importance of current know-how for the future of bioinspired materials? In this paper we intend to review and update the available methodologies to synthesize HAp/collagen composites, along with their characteristics. In addition, the future of these materials in terms of applications and their potential as a cutting-edge technology is discussed.
Collapse
Affiliation(s)
- Anabela Veiga
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal.,Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Filipa Castro
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Fernando Rocha
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Ana L Oliveira
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| |
Collapse
|
426
|
Zeimaran E, Pourshahrestani S, Fathi A, Razak NABA, Kadri NA, Sheikhi A, Baino F. Advances in bioactive glass-containing injectable hydrogel biomaterials for tissue regeneration. Acta Biomater 2021; 136:1-36. [PMID: 34562661 DOI: 10.1016/j.actbio.2021.09.034] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Successful tissue regeneration requires a scaffold with tailorable biodegradability, tissue-like mechanical properties, structural similarity to extracellular matrix (ECM), relevant bioactivity, and cytocompatibility. In recent years, injectable hydrogels have spurred increasing attention in translational medicine as a result of their tunable physicochemical properties in response to the surrounding environment. Furthermore, they have the potential to be implanted via minimally invasive procedures while enabling deep penetration, which is considered a feasible alternative to traditional open surgical procedures. However, polymeric hydrogels may lack sufficient stability and bioactivity in physiological environments. Composite hydrogels containing bioactive glass (BG) particulates, synergistically combining the advantages of their constituents, have emerged as multifunctional biomaterials with tailored mechanical properties and biological functionalities. This review paper highlights the recent advances in injectable composite hydrogel systems based on biodegradable polymers and BGs. The influence of BG particle geometry, composition, and concentration on gel formation, rheological and mechanical behavior as well as hydration and biodegradation of injectable hydrogels have been discussed. The applications of these composite hydrogels in tissue engineering are additionally described, with particular attention to bone and skin. Finally, the prospects and current challenges in the development of desirable injectable bioactive hydrogels for tissue regeneration are discussed to outline a roadmap for future research. STATEMENT OF SIGNIFICANCE: Developing a biomaterial that can be readily available for surgery, implantable via minimally invasive procedures, and be able to effectively stimulate tissue regeneration is one of the grand challenges in modern biomedicine. This review summarizes the state-of-the-art of injectable bioactive glass-polymer composite hydrogels to address several challenges in bone and soft tissue repair. The current limitations and the latest evolutions of these composite biomaterials are critically examined, and the roles of design parameters, such as composition, concentration, and size of the bioactive phase, and polymer-glass interactions on the rheological, mechanical, biological, and overall functional performance of hydrogels are detailed. Existing results and new horizons are discussed to provide a state-of-the-art review that may be useful for both experienced and early-stage researchers in the biomaterials community.
Collapse
|
427
|
In Vivo Efficacy of Neutrophil-Mediated Bone Regeneration Using a Rabbit Calvarial Defect Model. Int J Mol Sci 2021; 22:ijms222313016. [PMID: 34884821 PMCID: PMC8657540 DOI: 10.3390/ijms222313016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 01/08/2023] Open
Abstract
Reconstruction of bone due to surgical removal or disease-related bony defects is a clinical challenge. It is known that the immune system exerts positive immunomodulatory effects on tissue repair and regeneration. In this study, we evaluated the in vivo efficacy of autologous neutrophils on bone regeneration using a rabbit calvarial defect model. Methods: Twelve rabbits, each with two surgically created calvarial bone defects (10 mm diameter), were randomly divided into two groups; (i) single application of neutrophils (SA-NP) vs. SA-NP control, and (ii) repetitive application of neutrophils (RA-NP) vs. RA-NP control. The animals were euthanized at 4 and 8 weeks post-operatively and the treatment outcomes were evaluated by micro-computed tomography, histology, and histomorphometric analyses. Results: The micro-CT analysis showed a significantly higher bone volume fraction (bone volume/total volume) in the neutrophil-treated groups, i.e., median interquartile range (IQR) SA-NP (18) and RA-NP (24), compared with the untreated controls, i.e., SA-NP (7) and RA-NP (14) at 4 weeks (p < 0.05). Similarly, new bone area fraction (bone area/total area) was significantly higher in neutrophil-treated groups at 4 weeks (p < 0.05). Both SA-NP and RA-NP had a considerably higher bone volume and bone area at 8 weeks, although the difference was not statistically significant. In addition, immunohistochemical analysis at 8 weeks revealed a higher expression of osteocalcin in both SA-NP and RA-NP groups. Conclusions: The present study provides first hand evidence that autologous neutrophils may have a positive effect on promoting new bone formation. Future studies should be performed with a larger sample size in non-human primate models. If proven feasible, this new promising strategy could bring clinical benefits for bone defects to the field of oral and maxillofacial surgery.
Collapse
|
428
|
Stahl A, Yang YP. Regenerative Approaches for the Treatment of Large Bone Defects. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:539-547. [PMID: 33138705 PMCID: PMC8739850 DOI: 10.1089/ten.teb.2020.0281] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022]
Abstract
A variety of engineered materials have gained acceptance in orthopedic practice as substitutes for autologous bone grafts, although the regenerative efficacy of these engineered grafts is still limited compared with that of transplanted native tissues. For bone defects greater than 4-5 cm, however, common bone grafting procedures are insufficient and more complicated surgical interventions are required to repair and regenerate the damaged or missing bone. In this review, we describe current grafting materials and surgical techniques for the reconstruction of large bone defects, followed by tissue engineering (TE) efforts to develop improved therapies. Particular emphasis is placed on graft vascularization, because for both autologous bone and engineered alternatives, achieving adequate vascular development within the regenerating bone tissues remains a significant challenge in the context of large bone defects. To this end, TE and surgical strategies to induce development of a vasculature within bone grafts are discussed. Impact statement This review aims to present an accessible and thorough overview of current orthopedic surgical techniques as well as bone tissue engineering and vascularization strategies that might one day offer improvements to clinical therapies for the repair of large bone defects. We consider the lessons that clinical orthopedic reconstructive practices can contribute to the push toward engineered bone.
Collapse
Affiliation(s)
- Alexander Stahl
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
- Department of Chemistry, Stanford University, Stanford, California, USA
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
- Department of Materials Science and Engineering, and Stanford University, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
429
|
Applying extrusion-based 3D printing technique accelerates fabricating complex biphasic calcium phosphate-based scaffolds for bone tissue regeneration. J Adv Res 2021; 40:69-94. [PMID: 36100335 PMCID: PMC9481949 DOI: 10.1016/j.jare.2021.12.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/09/2021] [Accepted: 12/23/2021] [Indexed: 12/17/2022] Open
Abstract
Biphasic calcium phosphates offer a chemically similar biomaterial to the natural bone, which can significantly accelerate bone formation and reconstruction. Robocasting is a suitable technique to produce porous scaffolds supporting cell viability, proliferation, and differentiation. This review discusses materials and methods utilized for BCP robocasting, considering recent advancements and existing challenges in using additives for bioink preparation. Commercialization and marketing approach, in-vitro and in-vivo evaluations, biologic responses, and post-processing steps are also investigated. Possible strategies and opportunities for the use of BCP toward injured bone regeneration along with clinical applications are discussed. The study proposes that BCP possesses an acceptable level of bone substituting, considering its challenges and struggles.
Background Aim of review Key scientific concepts of review
Collapse
|
430
|
Porrelli D, Gruppuso M, Vecchies F, Marsich E, Turco G. Alginate bone scaffolds coated with a bioactive lactose modified chitosan for human dental pulp stem cells proliferation and differentiation. Carbohydr Polym 2021; 273:118610. [PMID: 34561009 DOI: 10.1016/j.carbpol.2021.118610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/21/2021] [Accepted: 08/20/2021] [Indexed: 12/27/2022]
Abstract
Bioactive and biodegradable porous scaffolds can hasten the healing of bone defects; moreover, patient stem cells seeded onto scaffolds can enhance the osteoinductive and osteoconductive properties of these biomaterials. In this work, porous alginate/hydroxyapatite scaffolds were functionalized with a bioactive coating of a lactose-modified chitosan (CTL). The highly interconnected porous structure of the scaffold was homogeneously coated with CTL. The scaffolds showed remarkable stability up to 60 days of aging. Human Dental Pulp Stem Cells (hDPSCs) cultured in the presence of CTL diluted in culture medium, showed a slight and negligible increase in terms of proliferation rate; on the contrary, an effect on osteogenic differentiation of the cells was observed as a significant increase in alkaline phosphatase activity. hDPSCs showed higher cell adhesion on CTL-coated scaffolds than on uncoated ones. CTL coating did not affect cell proliferation, but stimulated cell differentiation as shown by alkaline phosphatase activity analysis.
Collapse
Affiliation(s)
- Davide Porrelli
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34125 Trieste, Italy.
| | - Martina Gruppuso
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34125 Trieste, Italy.
| | - Federica Vecchies
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy.
| | - Eleonora Marsich
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Via Licio Giorgieri 5, 34129 Trieste, Italy.
| | - Gianluca Turco
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34125 Trieste, Italy.
| |
Collapse
|
431
|
Man K, Mekhileri NV, Lim KS, Jiang LH, Woodfield TBF, Yang XB. MI192 induced epigenetic reprogramming enhances the therapeutic efficacy of human bone marrows stromal cells for bone regeneration. Bone 2021; 153:116138. [PMID: 34339909 DOI: 10.1016/j.bone.2021.116138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022]
Abstract
Human bone marrow stromal cells (hBMSCs) have been extensively utilised for bone tissue engineering applications. However, they are associated with limitations that hinder their clinical utility for bone regeneration. Cell fate can be modulated via altering their epigenetic functionality. Inhibiting histone deacetylase (HDAC) enzymes have been reported to promote osteogenic differentiation, with HDAC3 activity shown to be causatively associated with osteogenesis. Therefore, this study aimed to investigate the potential of using an HDAC2 & 3 selective inhibitor - MI192 to induce epigenetic reprogramming of hBMSCs and enhance its therapeutic efficacy for bone formation. Treatment with MI192 caused a time-dose dependant reduction in hBMSCs viability. MI192 was also found to substantially alter hBMSCs epigenetic function through reduced HDAC activity and increased histone acetylation. hBMSCs were pre-treated with MI192 (50 μM) for 48 h prior to osteogenic induction. MI192 pre-treatment significantly upregulated osteoblast-related gene/protein expression (Runx2, ALP, Col1a and OCN) and enhanced alkaline phosphatase specific activity (ALPSA) (1.43-fold) (P ≤ 0.001). Moreover, MI192 substantially increased hBMSCs extracellular matrix calcium deposition (1.4-fold) (P ≤ 0.001) and mineralisation when compared to the untreated control. In 3D microtissue culture, MI192 significantly promoted hBMSCs osteoblast-related gene expression and ALPSA (> 2.41-fold) (P ≤ 0.001). Importantly, MI192 substantially enhanced extracellular matrix deposition (ALP, Col1a, OCN) and mineralisation (1.67-fold) (P ≤ 0.001) within the bioassembled-microtissue (BMT) construct. Following 8-week intraperitoneal implantation within nude mice, MI192 treated hBMSCs exhibited enhanced extracellular matrix deposition and mineralisation (2.39-fold) (P ≤ 0.001) within the BMT when compared to the untreated BMT construct. Taken together, these results demonstrate that MI192 effectively altered hBMSCs epigenetic functionality and is capable of promoting hBMSCs osteogenic differentiation in vitro and in vivo, indicating the potential of using epigenetic reprogramming to enhance the therapeutic efficacy of hBMSCs for bone augmentation strategies.
Collapse
Affiliation(s)
- Kenny Man
- Biomaterial and Tissue Engineering Group, School of Dentistry, University of Leeds, Leeds, UK; School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Naveen V Mekhileri
- CReaTE Group, Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Khoon S Lim
- CReaTE Group, Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Tim B F Woodfield
- CReaTE Group, Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Xuebin B Yang
- Biomaterial and Tissue Engineering Group, School of Dentistry, University of Leeds, Leeds, UK.
| |
Collapse
|
432
|
Subbiah R, Balbinot GDS, Athirasala A, Collares FM, Sereda G, Bertassoni LE. Nanoscale mineralization of cell-laden methacrylated gelatin hydrogels using calcium carbonate-calcium citrate core-shell microparticles. J Mater Chem B 2021; 9:9583-9593. [PMID: 34779469 DOI: 10.1039/d1tb01673c] [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: 11/21/2022]
Abstract
Conventional biomaterials developed for bone regeneration fail to fully recapitulate the nanoscale structural organization and complex composition of the native bone microenvironment. Therefore, despite promoting osteogenic differentiation of stem cells, they fall short of providing the structural, biochemical, and mechanical stimuli necessary to drive osteogenesis for bone regeneration and function. To address this, we have recently developed a novel strategy to engineer bone-like tissue using a biomimetic approach to achieve rapid and controlled nanoscale mineralization of a cell-laden matrix in the presence of osteopontin, a non-collagenous protein, and a supersaturated solution of calcium and phosphate medium. Here, we build on this approach to engineer bone regeneration scaffolds comprising methacrylated gelatin (GelMA) hydrogels incorporated with calcium citrate core-shell microparticles as a sustained and reliable source of calcium ions for in situ mineralization. We demonstrate successful biomineralization of GelMA hydrogels by embedded calcium carbonate-calcium citrate core-shell microparticles with the resultant mineral chemistry, structure, and organization reminiscent of that of native bone. The biomimetic mineralization was further shown to promote osteogenic differentiation of encapsulated human mesenchymal stem cells even in the absence of other exogenous osteogenic induction factors. Ultimately, by combining the superior biological response engendered by biomimetic mineralization with the intrinsic tissue engineering advantages offered by GelMA, such as biocompatibility, biodegradability, and printability, we envision that our system offers great potential for bone regeneration efforts.
Collapse
Affiliation(s)
- Ramesh Subbiah
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
| | - Gabriela de Souza Balbinot
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Dental Materials, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Avathamsa Athirasala
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
| | - Fabricio Mezzomo Collares
- Department of Dental Materials, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Grigoriy Sereda
- Department of Chemistry, University of South Dakota, Vermillion, SD 57069, USA.
| | - Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Center for Regenerative Medicine, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
| |
Collapse
|
433
|
Wei W, Dai H. Articular cartilage and osteochondral tissue engineering techniques: Recent advances and challenges. Bioact Mater 2021; 6:4830-4855. [PMID: 34136726 PMCID: PMC8175243 DOI: 10.1016/j.bioactmat.2021.05.011] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/20/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
In spite of the considerable achievements in the field of regenerative medicine in the past several decades, osteochondral defect regeneration remains a challenging issue among diseases in the musculoskeletal system because of the spatial complexity of osteochondral units in composition, structure and functions. In order to repair the hierarchical tissue involving different layers of articular cartilage, cartilage-bone interface and subchondral bone, traditional clinical treatments including palliative and reparative methods have showed certain improvement in pain relief and defect filling. It is the development of tissue engineering that has provided more promising results in regenerating neo-tissues with comparable compositional, structural and functional characteristics to the native osteochondral tissues. Here in this review, some basic knowledge of the osteochondral units including the anatomical structure and composition, the defect classification and clinical treatments will be first introduced. Then we will highlight the recent progress in osteochondral tissue engineering from perspectives of scaffold design, cell encapsulation and signaling factor incorporation including bioreactor application. Clinical products for osteochondral defect repair will be analyzed and summarized later. Moreover, we will discuss the current obstacles and future directions to regenerate the damaged osteochondral tissues.
Collapse
Affiliation(s)
- Wenying Wei
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan, 528200, China
| |
Collapse
|
434
|
McCarthy C, Camci-Unal G. Low Intensity Pulsed Ultrasound for Bone Tissue Engineering. MICROMACHINES 2021; 12:1488. [PMID: 34945337 PMCID: PMC8707172 DOI: 10.3390/mi12121488] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/16/2022]
Abstract
As explained by Wolff's law and the mechanostat hypothesis, mechanical stimulation can be used to promote bone formation. Low intensity pulsed ultrasound (LIPUS) is a source of mechanical stimulation that can activate the integrin/phosphatidylinositol 3-OH kinase/Akt pathway and upregulate osteogenic proteins through the production of cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2). This paper analyzes the results of in vitro and in vivo studies that have evaluated the effects of LIPUS on cell behavior within three-dimensional (3D) titanium, ceramic, and hydrogel scaffolds. We focus specifically on cell morphology and attachment, cell proliferation and viability, osteogenic differentiation, mineralization, bone volume, and osseointegration. As shown by upregulated levels of alkaline phosphatase and osteocalcin, increased mineral deposition, improved cell ingrowth, greater scaffold pore occupancy by bone tissue, and superior vascularization, LIPUS generally has a positive effect and promotes bone formation within engineered scaffolds. Additionally, LIPUS can have synergistic effects by producing the piezoelectric effect and enhancing the benefits of 3D hydrogel encapsulation, growth factor delivery, and scaffold modification. Additional research should be conducted to optimize the ultrasound parameters and evaluate the effects of LIPUS with other types of scaffold materials and cell types.
Collapse
Affiliation(s)
- Colleen McCarthy
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, MA 01854, USA;
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, MA 01854, USA;
- Department of Surgery, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01605, USA
| |
Collapse
|
435
|
Oliveira Pinho F, Pinto Joazeiro P, Santos AR. Evaluation of the Growth and Differentiation of Human Fetal Osteoblasts (hFOB) Cells on Demineralized Bone Matrix (DBM). Organogenesis 2021; 17:136-149. [PMID: 34845978 DOI: 10.1080/15476278.2021.2003134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Cells with osteogenic potential are believed to be an ideal source for bone tissue bioengineering. Large bone defects require temporary substitution of the damaged parts. In this respect, the transplantation of bone cells cultured on osteogenic substrates has been investigated. To use the natural bone matrix, one approach is the so-called demineralized bone matrix (DBM). In this study, we evaluated the interaction of human fetal osteoblasts (hFOB 1.19 cells, a human fetal osteoblastic cell line) with DBM fragments. No additional bone differentiation inducer was used other than the DBM itself. The samples were processed, had adhesion pattern evaluated and analyzed by light microscopy (cytochemical and immunocytochemical analysis) and electron microscopy (scanning and transmission). The adhesion pattern of hFOB cells on DBM was similar to what was observed on the cell culture plate. Morphological analysis showed that the hFOB cells had emitted filopodia and cellular projections on both controls and DBM. On DBM, the adhered cells emitted prolongations and migrated into the matrix. The monolayer growth pattern was observed as well as the accumulation of filamentous and reticulate extracellular materials when hFOB cells were cultured on the DBM surface. EDS analysis revealed the deposition of calcium on DBM. Immunocytochemical data showed that the hFOB cells were able to secrete extracellular matrix molecules such as fibronectin and laminin on DBM. Our data indicate that DBM successfully stimulates the osteoblastic phenotype of osteoblast-like cells and corroborate with the fact that DBM is a considerable natural matrix that promotes fractured-bone healing.
Collapse
Affiliation(s)
- Flavia Oliveira Pinho
- Departamento de Bioquímica e Biologia Tecidual, Instituto de Biologia, UNICAM, Campinas, SP Brazil
| | - Paulo Pinto Joazeiro
- Departamento de Bioquímica e Biologia Tecidual, Instituto de Biologia, UNICAM, Campinas, SP Brazil
| | - Arnaldo R Santos
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| |
Collapse
|
436
|
Huang G, Pan ST, Qiu JX. The osteogenic effects of porous Tantalum and Titanium alloy scaffolds with different unit cell structure. Colloids Surf B Biointerfaces 2021; 210:112229. [PMID: 34875470 DOI: 10.1016/j.colsurfb.2021.112229] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/30/2021] [Accepted: 11/15/2021] [Indexed: 12/28/2022]
Abstract
Porous scaffolds have long been regarded as optimal substitute for bone tissue repairing. In order to explore the influence of unit cell structure and inherent material characteristics on the porous scaffolds in terms of mechanical and biological performance, selective laser melting (SLM) technology was used to fabricate porous tantalum (Ta) and titanium alloy (Ti6Al4V) with diamond (Di) or rhombic dodecahedron (Do) unit cell structure. The mechanical strength of all the porous scaffolds could match that of trabecular bone, while the biological performance of each scaffold was diverse from each other. Moreover, the ILK/ERK1/2/Runx2 signaling pathway had been verified to be involved in the osteogenic differentiation of rat bone mesenchymal stem cells (rBMSCs) cultured on those porous scaffolds. Unit cell structure and material characteristics of the porous Ta and Ti6Al4V scaffolds can synergistically modulate this axis and further impact on the osteogenic effects. Our results hence illustrate that porous Ta scaffold with diamond unit cell structure possesses excellent osteogenic effects and moderate mechanical strength and porous Ti6Al4V scaffold with rhombic dodecahedron unit cell structure has the highest mechanical strength and moderate osteogenic effects. Both porous Ta and Ti6Al4V can be applied in different settings requiring either better biological performance or higher mechanical demand.
Collapse
Affiliation(s)
- Gan Huang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Shu-Ting Pan
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jia-Xuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
437
|
Lin C, Wang Y, Huang Z, Wu T, Xu W, Wu W, Xu Z. Advances in Filament Structure of 3D Bioprinted Biodegradable Bone Repair Scaffolds. Int J Bioprint 2021; 7:426. [PMID: 34805599 PMCID: PMC8600304 DOI: 10.18063/ijb.v7i4.426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/03/2021] [Indexed: 12/18/2022] Open
Abstract
Conventional bone repair scaffolds can no longer meet the high standards and requirements of clinical applications in terms of preparation process and service performance. Studies have shown that the diversity of filament structures of implantable scaffolds is closely related to their overall properties (mechanical properties, degradation properties, and biological properties). To better elucidate the characteristics and advantages of different filament structures, this paper retrieves and summarizes the state of the art in the filament structure of the three-dimensional (3D) bioprinted biodegradable bone repair scaffolds, mainly including single-layer structure, double-layer structure, hollow structure, core-shell structure and bionic structures. The eximious performance of the novel scaffolds was discussed from different aspects (material composition, ink configuration, printing parameters, etc.). Besides, the additional functions of the current bone repair scaffold, such as chondrogenesis, angiogenesis, anti-bacteria, and anti-tumor, were also concluded. Finally, the paper prospects the future material selection, structural design, functional development, and performance optimization of bone repair scaffolds.
Collapse
Affiliation(s)
- Chengxiong Lin
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China
| | - Yaocheng Wang
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China.,School of Railway Tracks and Transportation, Wuyi University, Jiangmen 529020, China
| | - Zhengyu Huang
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China.,School of Railway Tracks and Transportation, Wuyi University, Jiangmen 529020, China
| | - Tingting Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China
| | - Weikang Xu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China
| | - Wenming Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China
| | - Zhibiao Xu
- School of Railway Tracks and Transportation, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
438
|
Kennedy IW, Tsimbouri PM, Campsie P, Sood S, Childs PG, Reid S, Young PS, Meek DRM, Goodyear CS, Dalby MJ. Nanovibrational stimulation inhibits osteoclastogenesis and enhances osteogenesis in co-cultures. Sci Rep 2021; 11:22741. [PMID: 34815449 PMCID: PMC8611084 DOI: 10.1038/s41598-021-02139-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 11/02/2021] [Indexed: 11/23/2022] Open
Abstract
Models of bone remodelling could be useful in drug discovery, particularly if the model is one that replicates bone regeneration with reduction in osteoclast activity. Here we use nanovibrational stimulation to achieve this in a 3D co-culture of primary human osteoprogenitor and osteoclast progenitor cells. We show that 1000 Hz frequency, 40 nm amplitude vibration reduces osteoclast formation and activity in human mononuclear CD14+ blood cells. Additionally, this nanoscale vibration both enhances osteogenesis and reduces osteoclastogenesis in a co-culture of primary human bone marrow stromal cells and bone marrow hematopoietic cells. Further, we use metabolomics to identify Akt (protein kinase C) as a potential mediator. Akt is known to be involved in bone differentiation via transforming growth factor beta 1 (TGFβ1) and bone morphogenetic protein 2 (BMP2) and it has been implicated in reduced osteoclast activity via Guanine nucleotide-binding protein subunit α13 (Gα13). With further validation, our nanovibrational bioreactor could be used to help provide humanised 3D models for drug screening.
Collapse
Affiliation(s)
- Ian W Kennedy
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| | - P Monica Tsimbouri
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Paul Campsie
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow, G1 1QE, UK
| | - Shatakshi Sood
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University Place, University of Glasgow, Glasgow, G12 8TA, UK
| | - Peter G Childs
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow, G1 1QE, UK
| | - Stuart Reid
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow, G1 1QE, UK
| | - Peter S Young
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Dominic R M Meek
- Department of Trauma and Orthopaedics, Queen Elizabeth University Hospital, Glasgow, G51 4TF, UK
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University Place, University of Glasgow, Glasgow, G12 8TA, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
439
|
Nogueira LFB, Maniglia BC, Buchet R, Millán JL, Ciancaglini P, Bottini M, Ramos AP. Three-dimensional cell-laden collagen scaffolds: From biochemistry to bone bioengineering. J Biomed Mater Res B Appl Biomater 2021; 110:967-983. [PMID: 34793621 DOI: 10.1002/jbm.b.34967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/05/2021] [Accepted: 10/30/2021] [Indexed: 12/22/2022]
Abstract
The bones can be viewed as both an organ and a material. As an organ, the bones give structure to the body, facilitate skeletal movement, and provide protection to internal organs. As a material, the bones consist of a hybrid organic/inorganic three-dimensional (3D) matrix, composed mainly of collagen, noncollagenous proteins, and a calcium phosphate mineral phase, which is formed and regulated by the orchestrated action of a complex array of cells including chondrocytes, osteoblasts, osteocytes, and osteoclasts. The interactions between cells, proteins, and minerals are essential for the bone functions under physiological loading conditions, trauma, and fractures. The organization of the bone's organic and inorganic phases stands out for its mechanical and biological properties and has inspired materials research. The objective of this review is to fill the gaps between the physical and biological characteristics that must be achieved to fabricate scaffolds for bone tissue engineering with enhanced performance. We describe the organization of bone tissue highlighting the characteristics that have inspired the development of 3D cell-laden collagenous scaffolds aimed at replicating the mechanical and biological properties of bone after implantation. The role of noncollagenous macromolecules in the organization of the collagenous matrix and mineralization ability of entrapped cells has also been reviewed. Understanding the modulation of cell activity by the extracellular matrix will ultimately help to improve the biological performance of 3D cell-laden collagenous scaffolds used for bone regeneration and repair as well as for in vitro studies aimed at unravelling physiological and pathological processes occurring in the bone.
Collapse
Affiliation(s)
- Lucas Fabricio Bahia Nogueira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil.,Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Bianca C Maniglia
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil
| | - Rene Buchet
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Ana Paula Ramos
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil
| |
Collapse
|
440
|
Vasilyev AV, Kuznetsova VS, Bukharova TB, Osidak EO, Grigoriev TE, Zagoskin YD, Nedorubova IA, Domogatsky SP, Babichenko II, Zorina OA, Kutsev SI, Chvalun SN, Kulakov AA, Losev FF, Goldshtein DV. Osteoinductive Moldable and Curable Bone Substitutes Based on Collagen, BMP-2 and Highly Porous Polylactide Granules, or a Mix of HAP/β-TCP. Polymers (Basel) 2021; 13:polym13223974. [PMID: 34833275 PMCID: PMC8621266 DOI: 10.3390/polym13223974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 01/15/2023] Open
Abstract
In dentistry, maxillofacial surgery, traumatology, and orthopedics, there is a need to use osteoplastic materials that have not only osteoinductive and osteoconductive properties but are also convenient for use. In the study, compositions based on collagen hydrogel were developed. Polylactide granules (PLA) or a traditional bone graft, a mixture of hydroxyapatite and β-tricalcium phosphate (HAP/β-TCP), were used for gel filling to improve mechanical osteoconductive properties of compositions. The mechanical tests showed that collagen hydrogels filled with 12 wt% highly porous PLA granules (elastic modulus 373 ± 55 kPa) or 35 wt% HAP/β-TCP granules (elastic modulus 451 ± 32 kPa) had optimal manipulative properties. All composite components were cytocompatible. The cell’s viability was above 90%, and the components’ structure facilitated the cell’s surface adhesion. The bone morphogenetic protein-2 (BMP-2) provided osteoinductive composition properties. It was impregnated directly into the collagen hydrogel with the addition of fibronectin or inside porous PLA granules. The implantation of a collagen hydrogel with BMP-2 and PLA granules into a critical-size calvarial defect in rats led to the formation of the most significant volume of bone tissue: 61 ± 15%. It was almost 2.5 times more than in the groups where a collagen-fibronectin hydrogel with a mixture of HAP/β-TCP (25 ± 7%) or a fibronectin-free composition with porous PLA granules impregnated with BMP-2 (23 ± 8%) were used. Subcutaneous implantation of the compositions also showed their high biocompatibility and osteogenic potential in the absence of a bone environment. Thus, the collagen-fibronectin hydrogel with BMP-2 and PLA granules has optimal biocompatibility, osteogenic, and manipulative properties.
Collapse
Affiliation(s)
- Andrey Vyacheslavovich Vasilyev
- Research Centre for Medical Genetics, Moskvorechye st., 1, 115478 Moscow, Russia; (V.S.K.); (T.B.B.); (I.A.N.); (S.I.K.); (D.V.G.)
- Central Research Institute of Dental and Maxillofacial Surgery, Timur Frunze st., 16, 119021 Moscow, Russia; (I.I.B.); (O.A.Z.); (A.A.K.); (F.F.L.)
- Department of Pathological Anatomy, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya st., 117198 Moscow, Russia
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
- Correspondence:
| | - Valeriya Sergeevna Kuznetsova
- Research Centre for Medical Genetics, Moskvorechye st., 1, 115478 Moscow, Russia; (V.S.K.); (T.B.B.); (I.A.N.); (S.I.K.); (D.V.G.)
- Central Research Institute of Dental and Maxillofacial Surgery, Timur Frunze st., 16, 119021 Moscow, Russia; (I.I.B.); (O.A.Z.); (A.A.K.); (F.F.L.)
| | - Tatyana Borisovna Bukharova
- Research Centre for Medical Genetics, Moskvorechye st., 1, 115478 Moscow, Russia; (V.S.K.); (T.B.B.); (I.A.N.); (S.I.K.); (D.V.G.)
| | | | - Timofei Evgenevich Grigoriev
- NRC “Kurchatov Institute”, 1, Akademika Kurchatova pl, 123182 Moscow, Russia; (T.E.G.); (Y.D.Z.); (S.N.C.)
- Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, 141701 Moscow, Russia
| | - Yuriy Dmitrievich Zagoskin
- NRC “Kurchatov Institute”, 1, Akademika Kurchatova pl, 123182 Moscow, Russia; (T.E.G.); (Y.D.Z.); (S.N.C.)
| | - Irina Alekseevna Nedorubova
- Research Centre for Medical Genetics, Moskvorechye st., 1, 115478 Moscow, Russia; (V.S.K.); (T.B.B.); (I.A.N.); (S.I.K.); (D.V.G.)
| | - Sergey Petrovich Domogatsky
- Imtek Ltd., 3rd Cherepkovskaya st., 15a, 121552 Moscow, Russia; (E.O.O.); (S.P.D.)
- Federal State Budgetary Institution National Medical Research Center of Cardiology Ministry of Health of the Russian Federation, 3rd Cherepkovskaya st., 15a, 121552 Moscow, Russia
| | - Igor Ivanovich Babichenko
- Central Research Institute of Dental and Maxillofacial Surgery, Timur Frunze st., 16, 119021 Moscow, Russia; (I.I.B.); (O.A.Z.); (A.A.K.); (F.F.L.)
- Department of Pathological Anatomy, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya st., 117198 Moscow, Russia
| | - Oksana Aleksandrovna Zorina
- Central Research Institute of Dental and Maxillofacial Surgery, Timur Frunze st., 16, 119021 Moscow, Russia; (I.I.B.); (O.A.Z.); (A.A.K.); (F.F.L.)
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Sergey Ivanovich Kutsev
- Research Centre for Medical Genetics, Moskvorechye st., 1, 115478 Moscow, Russia; (V.S.K.); (T.B.B.); (I.A.N.); (S.I.K.); (D.V.G.)
| | - Sergei Nicolaevich Chvalun
- NRC “Kurchatov Institute”, 1, Akademika Kurchatova pl, 123182 Moscow, Russia; (T.E.G.); (Y.D.Z.); (S.N.C.)
| | - Anatoly Alekseevich Kulakov
- Central Research Institute of Dental and Maxillofacial Surgery, Timur Frunze st., 16, 119021 Moscow, Russia; (I.I.B.); (O.A.Z.); (A.A.K.); (F.F.L.)
| | - Fedor Fedorovich Losev
- Central Research Institute of Dental and Maxillofacial Surgery, Timur Frunze st., 16, 119021 Moscow, Russia; (I.I.B.); (O.A.Z.); (A.A.K.); (F.F.L.)
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Dmitry Vadimovich Goldshtein
- Research Centre for Medical Genetics, Moskvorechye st., 1, 115478 Moscow, Russia; (V.S.K.); (T.B.B.); (I.A.N.); (S.I.K.); (D.V.G.)
| |
Collapse
|
441
|
Pitta Kruize C, Panahkhahi S, Putra NE, Diaz-Payno P, van Osch G, Zadpoor AA, Mirzaali MJ. Biomimetic Approaches for the Design and Fabrication of Bone-to-Soft Tissue Interfaces. ACS Biomater Sci Eng 2021. [PMID: 34784181 DOI: 10.1021/acsbiomaterials.1c00620] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Bone-to-soft tissue interfaces are responsible for transferring loads between tissues with significantly dissimilar material properties. The examples of connective soft tissues are ligaments, tendons, and cartilages. Such natural tissue interfaces have unique microstructural properties and characteristics which avoid the abrupt transitions between two tissues and prevent formation of stress concentration at their connections. Here, we review some of the important characteristics of these natural interfaces. The native bone-to-soft tissue interfaces consist of several hierarchical levels which are formed in a highly specialized anisotropic fashion and are composed of different types of heterogeneously distributed cells. The characteristics of a natural interface can rely on two main design principles, namely by changing the local microarchitectural features (e.g., complex cell arrangements, and introducing interlocking mechanisms at the interfaces through various geometrical designs) and changing the local chemical compositions (e.g., a smooth and gradual transition in the level of mineralization). Implementing such design principles appears to be a promising approach that can be used in the design, reconstruction, and regeneration of engineered biomimetic tissue interfaces. Furthermore, prominent fabrication techniques such as additive manufacturing (AM) including 3D printing and electrospinning can be used to ease these implementation processes. Biomimetic interfaces have several biological applications, for example, to create synthetic scaffolds for osteochondral tissue repair.
Collapse
Affiliation(s)
- Carlos Pitta Kruize
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Sara Panahkhahi
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Niko Eka Putra
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Pedro Diaz-Payno
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Gerjo van Osch
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Mohammad J Mirzaali
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| |
Collapse
|
442
|
Hydrogel, Electrospun and Composite Materials for Bone/Cartilage and Neural Tissue Engineering. MATERIALS 2021; 14:ma14226899. [PMID: 34832300 PMCID: PMC8624846 DOI: 10.3390/ma14226899] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022]
Abstract
Injuries of the bone/cartilage and central nervous system are still a serious socio-economic problem. They are an effect of diversified, difficult-to-access tissue structures as well as complex regeneration mechanisms. Currently, commercially available materials partially solve this problem, but they do not fulfill all of the bone/cartilage and neural tissue engineering requirements such as mechanical properties, biochemical cues or adequate biodegradation. There are still many things to do to provide complete restoration of injured tissues. Recent reports in bone/cartilage and neural tissue engineering give high hopes in designing scaffolds for complete tissue regeneration. This review thoroughly discusses the advantages and disadvantages of currently available commercial scaffolds and sheds new light on the designing of novel polymeric scaffolds composed of hydrogels, electrospun nanofibers, or hydrogels loaded with nano-additives.
Collapse
|
443
|
Chen H, Shi Q, Shui H, Wang P, Chen Q, Li Z. Degradation of 3D-Printed Porous Polylactic Acid Scaffolds Under Mechanical Stimulus. Front Bioeng Biotechnol 2021; 9:691834. [PMID: 34765591 PMCID: PMC8576397 DOI: 10.3389/fbioe.2021.691834] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Polylactic acid (PLA) is a biodegradable polymer commonly used as a scaffold material to repair tissue defects, and its degradation is associated with mechanical stimulus. In this study, the effect of mechanical stimulus on the degradation of 3D-printed PLA scaffolds was investigated by in vitro experiments and an author-developed numerical model. Forty-five samples with porosity 64.8% were printed to carry out the degradation experiment within 90 days. Statistical analyses of the mass, volume fraction, Young’s modulus, and number average molecular weight were made, and the in vitro experiments were further used to verify the proposed numerical model of the scaffold degradation. The results indicated that the mechanical stimulus accelerated the degradation of the PLA scaffold, and the higher mechanical stimulus led to a faster degradation of the scaffolds at the late stage of the degradation process. In addition, the Young’s modulus and the normalized number average molecular weight of the PLA scaffolds between the experiments and the numerical simulations were comparable, especially for the number average molecular weight. The present study could be helpful in the design of the biodegradable PLA scaffolds.
Collapse
Affiliation(s)
- Heming Chen
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Quan Shi
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Hengtao Shui
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Peng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qiang Chen
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zhiyong Li
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China.,School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
444
|
Campodoni E, Velez M, Fragogeorgi E, Morales I, de la Presa P, Stanicki D, Dozio SM, Xanthopoulos S, Bouziotis P, Dermisiadou E, Rouchota M, Loudos G, Marín P, Laurent S, Boutry S, Panseri S, Montesi M, Tampieri A, Sandri M. Magnetic and radio-labeled bio-hybrid scaffolds to promote and track in vivo the progress of bone regeneration. Biomater Sci 2021; 9:7575-7590. [PMID: 34665185 DOI: 10.1039/d1bm00858g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
This work describes the preparation, characterization and functionalization with magnetic nanoparticles of a bone tissue-mimetic scaffold composed of collagen and hydroxyapatite obtained through a biomineralization process. Bone remodeling takes place over several weeks and the possibility to follow it in vivo in a quick and reliable way is still an outstanding issue. Therefore, this work aims to produce an implantable material that can be followed in vivo during bone regeneration by using the existing non-invasive imaging techniques (MRI). To this aim, suitably designed biocompatible SPIONs were linked to the hybrid scaffold using two different strategies, one involving naked SPIONs (nMNPs) and the other using coated and activated SPIONs (MNPs) exposing carboxylic acid functions allowing a covalent attachment between MNPs and collagen molecules. Physico-chemical characterization was carried out to investigate the morphology, crystallinity and stability of the functionalized materials followed by MRI analyses and evaluation of a radiotracer uptake ([99mTc]Tc-MDP). Cell proliferation assays in vitro were carried out to check the cytotoxicity and demonstrated no side effects due to the SPIONs. The achieved results demonstrated that the naked and coated SPIONs are more homogeneously distributed in the scaffold when incorporated during the synthesis process. This work demonstrated a suitable approach to develop a biomaterial for bone regeneration that allows the monitoring of the healing progress even for long-term follow-up studies.
Collapse
Affiliation(s)
- Elisabetta Campodoni
- Institute of Science and Technology for Ceramics-National Research Council (CNR), Faenza, Italy.
| | - Marisela Velez
- Instituto de Catálisis y Petroleoquímica (CSIC), Madrid, Spain.
| | - Eirini Fragogeorgi
- National Center for Scientific Research (NCSR) "Demokritos", Institute of Nuclear & Radiological Sciences & Technology, Energy &Safety, Ag. Paraskevi-Athens, Greece.,BIOEMTECH, Lefkippos Attica Technology Park, NCSR "Demokritos", Ag. Paraskevi-Athens, Greece
| | - Irene Morales
- Instituto de Magnetismo Aplicado (UCM-ADIF-CSIC), A6 22, Las Rozas, 28260, Spain.,Dpto Física de Materiales, UCM, Ciudad Universitaria, Madrid, 28040, Spain
| | - Patricia de la Presa
- Instituto de Magnetismo Aplicado (UCM-ADIF-CSIC), A6 22, Las Rozas, 28260, Spain.,Dpto Física de Materiales, UCM, Ciudad Universitaria, Madrid, 28040, Spain
| | - Dimitri Stanicki
- University of Mons, General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Lab, 7000 Mons, Belgium
| | - Samuele M Dozio
- Institute of Science and Technology for Ceramics-National Research Council (CNR), Faenza, Italy. .,Institute of Solid-State Electronics, Vienna University of Technology, Vienna, Austria
| | - Stavros Xanthopoulos
- National Center for Scientific Research (NCSR) "Demokritos", Institute of Nuclear & Radiological Sciences & Technology, Energy &Safety, Ag. Paraskevi-Athens, Greece
| | - Penelope Bouziotis
- National Center for Scientific Research (NCSR) "Demokritos", Institute of Nuclear & Radiological Sciences & Technology, Energy &Safety, Ag. Paraskevi-Athens, Greece
| | - Eleftheria Dermisiadou
- BIOEMTECH, Lefkippos Attica Technology Park, NCSR "Demokritos", Ag. Paraskevi-Athens, Greece
| | - Maritina Rouchota
- BIOEMTECH, Lefkippos Attica Technology Park, NCSR "Demokritos", Ag. Paraskevi-Athens, Greece
| | - George Loudos
- National Center for Scientific Research (NCSR) "Demokritos", Institute of Nuclear & Radiological Sciences & Technology, Energy &Safety, Ag. Paraskevi-Athens, Greece.,BIOEMTECH, Lefkippos Attica Technology Park, NCSR "Demokritos", Ag. Paraskevi-Athens, Greece
| | - Pilar Marín
- Instituto de Magnetismo Aplicado (UCM-ADIF-CSIC), A6 22, Las Rozas, 28260, Spain.,Dpto Física de Materiales, UCM, Ciudad Universitaria, Madrid, 28040, Spain
| | - Sophie Laurent
- University of Mons, General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Lab, 7000 Mons, Belgium.,Center for Microscopy and Molecular Imaging, 6041 Charleroi, Belgium
| | - Sébastien Boutry
- University of Mons, General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Lab, 7000 Mons, Belgium.,Center for Microscopy and Molecular Imaging, 6041 Charleroi, Belgium
| | - Silvia Panseri
- Institute of Science and Technology for Ceramics-National Research Council (CNR), Faenza, Italy.
| | - Monica Montesi
- Institute of Science and Technology for Ceramics-National Research Council (CNR), Faenza, Italy.
| | - Anna Tampieri
- Institute of Science and Technology for Ceramics-National Research Council (CNR), Faenza, Italy.
| | - Monica Sandri
- Institute of Science and Technology for Ceramics-National Research Council (CNR), Faenza, Italy.
| |
Collapse
|
445
|
Zhang J, Jiang Y, Shang Z, Zhao B, Jiao M, Liu W, Cheng M, Zhai B, Guo Y, Liu B, Shi X, Ma B. Biodegradable metals for bone defect repair: A systematic review and meta-analysis based on animal studies. Bioact Mater 2021; 6:4027-4052. [PMID: 33997491 PMCID: PMC8089787 DOI: 10.1016/j.bioactmat.2021.03.035] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 12/18/2022] Open
Abstract
Biodegradable metals are promising candidates for bone defect repair. With an evidence-based approach, this study investigated and analyzed the performance and degradation properties of biodegradable metals in animal models for bone defect repair to explore their potential clinical translation. Animal studies on bone defect repair with biodegradable metals in comparison with other traditional biomaterials were reviewed. Data was carefully collected after identification of population, intervention, comparison, outcome, and study design (PICOS), and following the inclusion criteria of biodegradable metals in animal studies. 30 publications on pure Mg, Mg alloys, pure Zn and Zn alloys were finally included after extraction from a collected database of 2543 publications. A qualitative systematic review and a quantitative meta-analysis were performed. Given the heterogeneity in animal model, anatomical site and critical size defect (CSD), biodegradable metals exhibited mixed effects on bone defect repair and degradation in animal studies in comparison with traditional non-degradable metals, biodegradable polymers, bioceramics, and autogenous bone grafts. The results indicated that there were limitations in the experimental design of the included studies, and quality of the evidence presented by the studies was very low. To enhance clinical translation of biodegradable metals, evidence-based research with data validity is needed. Future studies should adopt standardized experimental protocols in investigating the effects of biodegradable metals on bone defect repair with animal models.
Collapse
Affiliation(s)
- Jiazhen Zhang
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Yanbiao Jiang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Zhizhong Shang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Bing Zhao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Mingyue Jiao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Wenbo Liu
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Maobo Cheng
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Bao Zhai
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Yajuan Guo
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Bin Liu
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Xinli Shi
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Bin Ma
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
- Institute of Health Data Science, Lanzhou University, Lanzhou, 730000, PR China
| |
Collapse
|
446
|
Elkhoury K, Morsink M, Sanchez-Gonzalez L, Kahn C, Tamayol A, Arab-Tehrany E. Biofabrication of natural hydrogels for cardiac, neural, and bone Tissue engineering Applications. Bioact Mater 2021; 6:3904-3923. [PMID: 33997485 PMCID: PMC8080408 DOI: 10.1016/j.bioactmat.2021.03.040] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/05/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022] Open
Abstract
Natural hydrogels are one of the most promising biomaterials for tissue engineering applications, due to their biocompatibility, biodegradability, and extracellular matrix mimicking ability. To surpass the limitations of conventional fabrication techniques and to recapitulate the complex architecture of native tissue structure, natural hydrogels are being constructed using novel biofabrication strategies, such as textile techniques and three-dimensional bioprinting. These innovative techniques play an enormous role in the development of advanced scaffolds for various tissue engineering applications. The progress, advantages, and shortcomings of the emerging biofabrication techniques are highlighted in this review. Additionally, the novel applications of biofabricated natural hydrogels in cardiac, neural, and bone tissue engineering are discussed as well.
Collapse
Affiliation(s)
| | - Margaretha Morsink
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7500AE, the Netherlands
| | | | - Cyril Kahn
- LIBio, Université de Lorraine, Nancy, F-54000, France
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT, 06030, USA
| | | |
Collapse
|
447
|
Hu J, Wang Z, Miszuk JM, Zhu M, Lansakara TI, Tivanski AV, Banas JA, Sun H. Vanillin-bioglass cross-linked 3D porous chitosan scaffolds with strong osteopromotive and antibacterial abilities for bone tissue engineering. Carbohydr Polym 2021; 271:118440. [PMID: 34364578 PMCID: PMC8353169 DOI: 10.1016/j.carbpol.2021.118440] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/21/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
Chitosan scaffolds crosslinked by current methods insufficiently meet the demands of bone tissue engineering applications. We developed a novel effective crosslinking technique by using the natural and safe vanillin together with bioglass microparticles to generate an antibacterial, osteoconductive, and mechanically robust 3D porous chitosan-vanillin-bioglass (CVB) scaffold. In addition to the significantly improved mechanical properties, the CVB scaffolds had high porosity (>90%) and interconnected macroporous structures. Our data suggested that the crosslinking mainly resulted from the Schiff base reactions between the aldehydes of vanillin and amines of chitosan, together with the hydrogen and ionic bonds formed within them. Importantly, the CVB scaffolds not only showed good biocompatibility, bioactivity, and strong antibacterial ability but also significantly promoted osteoblastic differentiation, mineralization in vitro, and ectopic bone formation in vivo. Thus, the CVB scaffolds hold great promise for bone tissue engineering applications based on their robust mechanical properties, osteoconductivity, and antibacterial abilities.
Collapse
Affiliation(s)
- Jue Hu
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, Iowa City, IA 52242, USA; Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Zhuozhi Wang
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, Iowa City, IA 52242, USA; Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Jacob M Miszuk
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, Iowa City, IA 52242, USA; Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Min Zhu
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | | | - Alexei V Tivanski
- Department of Chemistry, University of Iowa, Iowa City, IA 52242, USA
| | - Jeffrey A Banas
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Hongli Sun
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, Iowa City, IA 52242, USA; Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA.
| |
Collapse
|
448
|
Jaganathan SK, Mani MP. Investigation of attributes of bourbon oil and cobalt nitrate constituted electrospun nanoscaffolds for blood compatibility and in vitro bone formation. AN ACAD BRAS CIENC 2021; 93:e20201140. [PMID: 34705943 DOI: 10.1590/0001-3765202120201140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 12/16/2020] [Indexed: 11/22/2022] Open
Abstract
This work aims to fabricate scaffold using polyurethane (PU) integrated with bourbon oil (BB) and cobalt nitrate (CoNO3) using the electrospinning technique. Morphological investigation signified a fall in fibre diameter for the PU/BB and PU/BB/CoNO3 nanocomposite than the PU. Spectral analysis indicated that BB and CoNO3 were added within the PU matrix. Wettability analysis insinuated an increase in the hydrophobic nature of the PU/BB than the PU. PU/BB/CoNO3 turned to be hydrophilic due to the integration of CoNO3 in the polymer matrix. Mechanical testing of PU/BB and PU/BB/CoNO3 indicated an increase in the tensile strength of the fabricated composites. Atomic force microscopy (AFM) portrayed the reduction in the roughness of the PU/BB and PU/BB/CoNO3 compared to the PU. The coagulation studies invariably documented the improved anticoagulant behaviour and less toxic nature of the PU/BB and PU/BB/CoNO3 in comparison with the PU. Further, bone mineralization testing revealed the enhanced apatite formation of the nanocomposite. Nanocomposite scaffolds with the fore-mentioned properties hold good potential for bone tissue engineering.
Collapse
Affiliation(s)
- Saravana K Jaganathan
- University of Hull, Department of Engineering, Faculty of Science and Engineering, HU6 7RX, Hull, U.K.,Universiti Teknologi Malaysia, School of Electrical Engineering, Faculty of Engineering, 81310, Johor Bahru, Malaysia
| | - Mohan P Mani
- Universiti Teknologi Malaysia, School of Biomedical Engineering and Health Sciences, Faculty of Engineering, 81310, Skudai, Malaysia
| |
Collapse
|
449
|
Man K, Brunet MY, Louth S, Robinson TE, Fernandez-Rhodes M, Williams S, Federici AS, Davies OG, Hoey DA, Cox SC. Development of a Bone-Mimetic 3D Printed Ti6Al4V Scaffold to Enhance Osteoblast-Derived Extracellular Vesicles' Therapeutic Efficacy for Bone Regeneration. Front Bioeng Biotechnol 2021; 9:757220. [PMID: 34765595 PMCID: PMC8576375 DOI: 10.3389/fbioe.2021.757220] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular Vesicles (EVs) are considered promising nanoscale therapeutics for bone regeneration. To date, EVs are typically procured from cells on 2D tissue culture plastic, an artificial environment that limits cell growth and does not replicate in situ biochemical or biophysical conditions. This study investigated the potential of 3D printed titanium scaffolds coated with hydroxyapatite to promote the therapeutic efficacy of osteoblast-derived EVs. Ti6Al4V titanium scaffolds with different pore sizes (500 and 1000 µm) and shapes (square and triangle) were fabricated by selective laser melting. A bone-mimetic nano-needle hydroxyapatite (nnHA) coating was then applied. EVs were procured from scaffold-cultured osteoblasts over 2 weeks and vesicle concentration was determined using the CD63 ELISA. Osteogenic differentiation of human bone marrow stromal cells (hBMSCs) following treatment with primed EVs was evaluated by assessing alkaline phosphatase activity, collagen production and calcium deposition. Triangle pore scaffolds significantly increased osteoblast mineralisation (1.5-fold) when compared to square architectures (P ≤ 0.001). Interestingly, EV yield was also significantly enhanced on these higher permeability structures (P ≤ 0.001), in particular (2.2-fold) for the larger pore structures (1000 µm). Furthermore osteoblast-derived EVs isolated from triangular pore scaffolds significantly increased hBMSCs mineralisation when compared to EVs acquired from square pore scaffolds (1.7-fold) and 2D culture (2.2-fold) (P ≤ 0.001). Coating with nnHA significantly improved osteoblast mineralisation (>2.6-fold) and EV production (4.5-fold) when compared to uncoated scaffolds (P ≤ 0.001). Together, these findings demonstrate the potential of harnessing bone-mimetic culture platforms to enhance the production of pro-regenerative EVs as an acellular tool for bone repair.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Mathieu Y. Brunet
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Sophie Louth
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Thomas E. Robinson
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Maria Fernandez-Rhodes
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Soraya Williams
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Angelica S. Federici
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin and RCSI, Dublin, Ireland
| | - Owen G. Davies
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - David A. Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin and RCSI, Dublin, Ireland
| | - Sophie C. Cox
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
450
|
Chitosan Covalently Functionalized with Peptides Mapped on Vitronectin and BMP-2 for Bone Tissue Engineering. NANOMATERIALS 2021; 11:nano11112784. [PMID: 34835549 PMCID: PMC8622029 DOI: 10.3390/nano11112784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/27/2022]
Abstract
Worldwide, over 20 million patients suffer from bone disorders annually. Bone scaffolds are designed to integrate into host tissue without causing adverse reactions. Recently, chitosan, an easily available natural polymer, has been considered a suitable scaffold for bone tissue growth as it is a biocompatible, biodegradable, and non-toxic material with antimicrobial activity and osteoinductive capacity. In this work, chitosan was covalently and selectively biofunctionalized with two suitably designed bioactive synthetic peptides: a Vitronectin sequence (HVP) and a BMP-2 peptide (GBMP1a). Nuclear magnetic resonance (NMR), X-ray photoelectron spectroscopy (XPS), and Fourier transform infrared spectroscopy (FT-IR) investigations highlighted the presence of the peptides grafted to chitosan (named Chit-HVP and Chit-GBMP1a). Chit-HVP and Chit-GBMP1a porous scaffolds promoted human osteoblasts adhesion, proliferation, calcium deposition, and gene expression of three crucial osteoblast proteins. In particular, Chit-HVP highly promoted adhesion and proliferation of osteoblasts, while Chit-GBMP1a guided cell differentiation towards osteoblastic phenotype.
Collapse
|