1
|
Amini-Mosleh-Abadi S, Yazdanpanah Z, Ketabat F, Saadatifar M, Mohammadi M, Salimi N, Asef Nejhad A, Sadeghianmaryan A. In vitro characterization of 3D printed polycaprolactone/graphene oxide scaffolds impregnated with alginate and gelatin hydrogels for bone tissue engineering. J Biomater Appl 2025:8853282251336552. [PMID: 40278887 DOI: 10.1177/08853282251336552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
To achieve successful bone tissue engineering (BTE), it is necessary to fabricate a biomedical scaffold with appropriate structure as well as favorable composition. Despite a broad range of studies, this remains a challenge, highlighting the need for a better understanding of how structural features (e.g., pore size) and scaffold composition influence mechanical and physical properties, as well as cellular behavior. Therefore, the objective of this study was to characterize physical properties (swelling, degradation), mechanical properties (compressive modulus), and cellular behavior in relation to varying compositions (referred to composite and hybrid scaffolds) as well as varying pore sizes in three-dimensional (3D) printed scaffolds. Composite scaffolds were fabricated from polycaprolactone (PCL) and two different graphene oxide (GO) (3% and 9% (w/v)) concentrations. Additionally, hybrid scaffolds were fabricated by impregnating 3D printed scaffolds in a hydrogel blend of alginate/gelatin. Pore sizes of 400, 1000, and 1500 μm were investigated in this study to assess their effect on the scaffold properties. Our findings showed that swelling and degradation properties were enhanced by (I) the addition of GO as well as introduction of both hydrogel and highest concentration of GO (9% (w/v) GO) into the polymeric matrix of PCL, and (II) increasing the pore size within the scaffolds. Mechanical testing revealed that compressive elastic modulus increased with decreasing pore size, incorporation of GO, and increasing GO content into the matrix of PCL. Although our investigated scaffolds with various pore sizes did not show comparable elastic moduli to that of cortical bone, they exhibited an elastic modulus range (∼31-48 MPa) matching that of trabecular bone. The highest compressive modulus (∼48 MPa) was observed in scaffolds of PCL/9% (w/v) GO (composite scaffolds) with the pore size of 400 μm. Cell viability assay demonstrated high MG-63 cell survival (greater than 70%) in all composite and hybrid scaffolds (indicating scaffold biocompatibility) except PCL/3% (w/v) GO scaffolds. The findings of this study contribute to the field of BTE by providing scaffold design insights in terms of pore size and composition.
Collapse
Affiliation(s)
| | - Zahra Yazdanpanah
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Farinaz Ketabat
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mahya Saadatifar
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Mohammadi
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nima Salimi
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azade Asef Nejhad
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ali Sadeghianmaryan
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Former Postdoctoral Research Fellow, Department of Biomedical Engineering, University of Memphis, Memphis, TN, USA
| |
Collapse
|
2
|
Golebiowska AA, Intravaia JT, Sathe V, Kumbar SG, Nukavarapu SP. Engineered Osteochondral Scaffolds with Bioactive Cartilage Zone for Enhanced Articular Cartilage Regeneration. Ann Biomed Eng 2025; 53:597-611. [PMID: 39602036 PMCID: PMC11835937 DOI: 10.1007/s10439-024-03655-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Despite progress, osteochondral (OC) tissue engineering strategies face limitations in terms of articular cartilage layer development and its integration with the underlying bone tissue. The main objective of this study is to develop a zonal OC scaffold with native biochemical signaling in the cartilage zone to promote articular cartilage development devoid of cells and growth factors. Herein, we report the development and in vivo assessment of a novel gradient and zonal-structured scaffold for OC defect regeneration. The scaffold system is composed of a mechanically supportive 3D-printed template containing decellularized cartilage extracellular matrix (ECM) biomaterial in the cartilage zone that possesses bioactive characteristics, such as chemotactic activity and native tissue biochemical composition. OC scaffolds with a bioactive cartilage zone were implanted in vivo in a rabbit osteochondral defect model and assessed for gross morphology, matrix deposition, cellular distribution, and overall tissue regeneration. The scaffold system supported recruitment and infiltration of host cells into the cartilage zone of the graft, which led to increased ECM deposition and physiologically relevant articular cartilage tissue formation. Semi-quantitative ICRS scoring (overall score double for OC scaffold with bioactive cartilage zone compared to PLA scaffold) further confirm the bioactive scaffold enhanced articular cartilage engineering. This strategy of designing bioactive scaffolds to promote endogenous cellular infiltration can be a much simpler and effective approach for OC tissue repair and regeneration.
Collapse
Affiliation(s)
- Aleksandra A Golebiowska
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
| | - Jonathon T Intravaia
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
| | - Vinayak Sathe
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Sangamesh G Kumbar
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Syam P Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA.
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA.
| |
Collapse
|
3
|
Da Silva K, Kumar P, Choonara YE. The paradigm of stem cell secretome in tissue repair and regeneration: Present and future perspectives. Wound Repair Regen 2025; 33:e13251. [PMID: 39780313 PMCID: PMC11711308 DOI: 10.1111/wrr.13251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
As the number of patients requiring organ transplants continues to rise exponentially, there is a dire need for therapeutics, with repair and regenerative properties, to assist in alleviating this medical crisis. Over the past decade, there has been a shift from conventional stem cell treatments towards the use of the secretome, the protein and factor secretions from cells. These components may possess novel druggable targets and hold the key to profoundly altering the field of regenerative medicine. Despite the progress in this field, clinical translation of secretome-containing products is limited by several challenges including but not limited to ensuring batch-to-batch consistency, the prevention of further heterogeneity, production of sufficient secretome quantities, product registration, good manufacturing practice protocols and the pharmacokinetic/pharmacodynamic profiles of all the components. Despite this, the secretome may hold the key to unlocking the regenerative blockage scientists have encountered for years. This review critically analyses the secretome derived from different cell sources and used in several tissues for tissue regeneration. Furthermore, it provides an overview of the current delivery strategies and the future perspectives for the secretome as a potential therapeutic. The success and possible shortcomings of the secretome are evaluated.
Collapse
Affiliation(s)
- Kate Da Silva
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
4
|
Wu H, Wang X, Wang G, Yuan G, Jia W, Tian L, Zheng Y, Ding W, Pei J. Advancing Scaffold-Assisted Modality for In Situ Osteochondral Regeneration: A Shift From Biodegradable to Bioadaptable. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407040. [PMID: 39104283 DOI: 10.1002/adma.202407040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/10/2024] [Indexed: 08/07/2024]
Abstract
Over the decades, the management of osteochondral lesions remains a significant yet unmet medical challenge without curative solutions to date. Owing to the complex nature of osteochondral units with multi-tissues and multicellularity, and inherently divergent cellular turnover capacities, current clinical practices often fall short of robust and satisfactory repair efficacy. Alternative strategies, particularly tissue engineering assisted with biomaterial scaffolds, achieve considerable advances, with the emerging pursuit of a more cost-effective approach of in situ osteochondral regeneration, as evolving toward cell-free modalities. By leveraging endogenous cell sources and innate regenerative potential facilitated with instructive scaffolds, promising results are anticipated and being evidenced. Accordingly, a paradigm shift is occurring in scaffold development, from biodegradable and biocompatible to bioadaptable in spatiotemporal control. Hence, this review summarizes the ongoing progress in deploying bioadaptable criteria for scaffold-based engineering in endogenous osteochondral repair, with emphases on precise control over the scaffolding material, degradation, structure and biomechanics, and surface and biointerfacial characteristics, alongside their distinguished impact on the outcomes. Future outlooks of a highlight on advanced, frontier materials, technologies, and tools tailoring precision medicine and smart healthcare are provided, which potentially paves the path toward the ultimate goal of complete osteochondral regeneration with function restoration.
Collapse
Affiliation(s)
- Han Wu
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite & Center of Hydrogen Science, School of Materials Science & Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xuejing Wang
- Interdisciplinary Research Center of Biology & Catalysis, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Guocheng Wang
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong, 518055, China
| | - Guangyin Yuan
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite & Center of Hydrogen Science, School of Materials Science & Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Weitao Jia
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Liangfei Tian
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Wenjiang Ding
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite & Center of Hydrogen Science, School of Materials Science & Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jia Pei
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite & Center of Hydrogen Science, School of Materials Science & Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Institute of Medical Robotics & National Engineering Research Center for Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
5
|
Desai N, Pande S, Vora L, Kommineni N. Correction to "Nanofibrous Microspheres: A Biomimetic Platform for Bone Tissue Regeneration". ACS APPLIED BIO MATERIALS 2024; 7:6325-6331. [PMID: 39162584 PMCID: PMC11409221 DOI: 10.1021/acsabm.4c01057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Indexed: 08/21/2024]
|
6
|
Desai N, Pande S, Vora LK, Kommineni N. Nanofibrous Microspheres: A Biomimetic Platform for Bone Tissue Regeneration. ACS APPLIED BIO MATERIALS 2024; 7:4270-4292. [PMID: 38950103 PMCID: PMC11253102 DOI: 10.1021/acsabm.4c00613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Bone, a fundamental constituent of the human body, is a vital scaffold for support, protection, and locomotion, underscoring its pivotal role in maintaining skeletal integrity and overall functionality. However, factors such as trauma, disease, or aging can compromise bone structure, necessitating effective strategies for regeneration. Traditional approaches often lack biomimetic environments conducive to efficient tissue repair. Nanofibrous microspheres (NFMS) present a promising biomimetic platform for bone regeneration by mimicking the native extracellular matrix architecture. Through optimized fabrication techniques and the incorporation of active biomolecular components, NFMS can precisely replicate the nanostructure and biochemical cues essential for osteogenesis promotion. Furthermore, NFMS exhibit versatile properties, including tunable morphology, mechanical strength, and controlled release kinetics, augmenting their suitability for tailored bone tissue engineering applications. NFMS enhance cell recruitment, attachment, and proliferation, while promoting osteogenic differentiation and mineralization, thereby accelerating bone healing. This review highlights the pivotal role of NFMS in bone tissue engineering, elucidating their design principles and key attributes. By examining recent preclinical applications, we assess their current clinical status and discuss critical considerations for potential clinical translation. This review offers crucial insights for researchers at the intersection of biomaterials and tissue engineering, highlighting developments in this expanding field.
Collapse
Affiliation(s)
- Nimeet Desai
- Department
of Biomedical Engineering, Indian Institute
of Technology Hyderabad, Kandi 502285, India
| | - Shreya Pande
- Department
of Biomedical Engineering, Indian Institute
of Technology Hyderabad, Kandi 502285, India
| | - Lalitkumar K. Vora
- School
of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Nagavendra Kommineni
- Center
for Biomedical Research, Population Council, New York, New York 10065, United States
| |
Collapse
|
7
|
Pecorini G, Braccini S, Simoni S, Corti A, Parrini G, Puppi D. Additive Manufacturing of Wet-Spun Poly(3-hydroxybutyrate-co-3-hydroxyvalerate)-Based Scaffolds Loaded with Hydroxyapatite. Macromol Biosci 2024; 24:e2300538. [PMID: 38534197 DOI: 10.1002/mabi.202300538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/20/2024] [Indexed: 03/28/2024]
Abstract
Tissue engineering represents an advanced therapeutic approach for the treatment of bone tissue defects. Polyhydroxyalkanoates are a promising class of natural polymers in this context thanks to their biocompatibility, processing versatility, and mechanical properties. The aim of this study is the development by computer-aided wet-spinning of novel poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV)-based composite scaffolds for bone engineering. In particular, PHBV scaffolds are loaded with hydroxyapatite (HA), an osteoinductive ceramic, in order to tailor their biological activity and mechanical properties. PHBV blending with poly(lactide-co-glycolide) (PLGA) is also explored to increase the processing properties of the polymeric mixture used for composite scaffold fabrication. Different HA percentages, up to 15% wt., can be loaded into the PHBV or PHBV/PLGA scaffolds without compromising their interconnected porous architecture, as well as the polymer morphological and thermal properties, as demonstrated by scanning electron microscopy, thermogravimetric analysis, and differential scanning calorimetry. In addition, HA loading results in increased scaffold compressive stiffness to levels comparable to those of trabecular bone tissue, as well as in higher in vitro MC3T3-E1 cell viability and production of mineralized extracellular matrix, in comparison to what observed for unloaded scaffolds. The observed mechanical and biological properties suggest the suitability of the developed scaffolds for bone engineering.
Collapse
Affiliation(s)
- Gianni Pecorini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | - Simona Braccini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | - Stefano Simoni
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | - Andrea Corti
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | | | - Dario Puppi
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| |
Collapse
|
8
|
Alizadeh S, Mahboobi L, Nasiri M, Khosrowpour Z, Khosravimelal S, Asgari F, Gholipour-Malekabadi M, Taghi Razavi-Toosi SM, Singh Chauhan NP, Ghobadi F, Nasiri H, Gholipourmalekabadi M. Decellularized Placental Sponge Seeded with Human Mesenchymal Stem Cells Improves Deep Skin Wound Healing in the Animal Model. ACS APPLIED BIO MATERIALS 2024; 7:2140-2152. [PMID: 38470456 DOI: 10.1021/acsabm.3c00747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Skin injuries lead to a large burden of morbidity. Although numerous clinical and scientific strategies have been investigated to repair injured skin, optimal regeneration therapy still poses a considerable obstacle. To address this challenge, decellularized extracellular matrix-based scaffolds recellularized with stem cells offer significant advancements in skin regeneration and wound healing. Herein, a decellularized human placental sponge (DPS) was fabricated using the decellularization and freeze-drying technique and then recellularized with human adipose-derived mesenchymal cells (MSCs). The biological and biomechanical properties and skin full-thickness wound healing capacity of the stem cells-DPS constructs were investigated in vitro and in vivo. The DPS exhibited a uniform 3D microstructure with an interconnected pore network, 89.21% porosity, a low degradation rate, and good mechanical properties. The DPS and MSCs-DPS constructs were implanted in skin full-thickness wound models in mice. An accelerated wound healing was observed in the wounds implanted with the MSCs-DPS construct when compared to DPS and control (wounds with no treatment) during 7 and 21 days postimplantation follow-up. In the MSCs-DPS group, the wound was completely re-epithelialized, the epidermis layer was properly organized, and the dermis and epidermis' bilayer structures were restored after 7 days. Our findings suggest that DPS is an excellent carrier for MSC culture and delivery to skin wounds and now promises to proceed with clinical evaluations.
Collapse
Affiliation(s)
- Sanaz Alizadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Leila Mahboobi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Modara Nasiri
- Department of Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran 19585, Iran
- Research Company Located in Islamic Azad University Science and Technology Park, Araz Fidar Azma, Tehran, 1477893855, Iran
| | - Zahra Khosrowpour
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Sadjad Khosravimelal
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Fatemeh Asgari
- Avicenna Infertility Clinic, Avicenna Research Institute, ACECR, Tehran 1985743413, Iran
| | | | - Seyyed Mohammad Taghi Razavi-Toosi
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht 41887-94755, Iran
- Medical Biotechnology Research Center, Guilan University of Medical Sciences, Rasht 41887-94755, Iran
| | - Narendra Pal Singh Chauhan
- Department of Chemistry, Faculty of Science, Bhupal Nobles' University, Udaipur, Rajasthan 313001, India
| | - Faezeh Ghobadi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran 14588-89694, Iran
| | - Hajar Nasiri
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| |
Collapse
|
9
|
Golebiowska AA, Intravaia JT, Sathe VM, Kumbar SG, Nukavarapu SP. Decellularized extracellular matrix biomaterials for regenerative therapies: Advances, challenges and clinical prospects. Bioact Mater 2024; 32:98-123. [PMID: 37927899 PMCID: PMC10622743 DOI: 10.1016/j.bioactmat.2023.09.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Tissue engineering and regenerative medicine have shown potential in the repair and regeneration of tissues and organs via the use of engineered biomaterials and scaffolds. However, current constructs face limitations in replicating the intricate native microenvironment and achieving optimal regenerative capacity and functional recovery. To address these challenges, the utilization of decellularized tissues and cell-derived extracellular matrix (ECM) has emerged as a promising approach. These biocompatible and bioactive biomaterials can be engineered into porous scaffolds and grafts that mimic the structural and compositional aspects of the native tissue or organ microenvironment, both in vitro and in vivo. Bioactive dECM materials provide a unique tissue-specific microenvironment that can regulate and guide cellular processes, thereby enhancing regenerative therapies. In this review, we explore the emerging frontiers of decellularized tissue-derived and cell-derived biomaterials and bio-inks in the field of tissue engineering and regenerative medicine. We discuss the need for further improvements in decellularization methods and techniques to retain structural, biological, and physicochemical characteristics of the dECM products in a way to mimic native tissues and organs. This article underscores the potential of dECM biomaterials to stimulate in situ tissue repair through chemotactic effects for the development of growth factor and cell-free tissue engineering strategies. The article also identifies the challenges and opportunities in developing sterilization and preservation methods applicable for decellularized biomaterials and grafts and their translation into clinical products.
Collapse
Affiliation(s)
| | - Jonathon T. Intravaia
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Vinayak M. Sathe
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Syam P. Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| |
Collapse
|
10
|
Mishchenko O, Yanovska A, Kosinov O, Maksymov D, Moskalenko R, Ramanavicius A, Pogorielov M. Synthetic Calcium-Phosphate Materials for Bone Grafting. Polymers (Basel) 2023; 15:3822. [PMID: 37765676 PMCID: PMC10536599 DOI: 10.3390/polym15183822] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Synthetic bone grafting materials play a significant role in various medical applications involving bone regeneration and repair. Their ability to mimic the properties of natural bone and promote the healing process has contributed to their growing relevance. While calcium-phosphates and their composites with various polymers and biopolymers are widely used in clinical and experimental research, the diverse range of available polymer-based materials poses challenges in selecting the most suitable grafts for successful bone repair. This review aims to address the fundamental issues of bone biology and regeneration while providing a clear perspective on the principles guiding the development of synthetic materials. In this study, we delve into the basic principles underlying the creation of synthetic bone composites and explore the mechanisms of formation for biologically important complexes and structures associated with the various constituent parts of these materials. Additionally, we offer comprehensive information on the application of biologically active substances to enhance the properties and bioactivity of synthetic bone grafting materials. By presenting these insights, our review enables a deeper understanding of the regeneration processes facilitated by the application of synthetic bone composites.
Collapse
Affiliation(s)
- Oleg Mishchenko
- Department of Surgical and Propaedeutic Dentistry, Zaporizhzhia State Medical and Pharmaceutical University, 26, Prosp. Mayakovskogo, 69035 Zaporizhzhia, Ukraine; (O.M.); (O.K.); (D.M.)
| | - Anna Yanovska
- Theoretical and Applied Chemistry Department, Sumy State University, R-Korsakova Street, 40007 Sumy, Ukraine
| | - Oleksii Kosinov
- Department of Surgical and Propaedeutic Dentistry, Zaporizhzhia State Medical and Pharmaceutical University, 26, Prosp. Mayakovskogo, 69035 Zaporizhzhia, Ukraine; (O.M.); (O.K.); (D.M.)
| | - Denys Maksymov
- Department of Surgical and Propaedeutic Dentistry, Zaporizhzhia State Medical and Pharmaceutical University, 26, Prosp. Mayakovskogo, 69035 Zaporizhzhia, Ukraine; (O.M.); (O.K.); (D.M.)
| | - Roman Moskalenko
- Department of Pathology, Sumy State University, R-Korsakova Street, 40007 Sumy, Ukraine;
| | - Arunas Ramanavicius
- NanoTechnas-Center of Nanotechnology and Materials Science, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania
| | - Maksym Pogorielov
- Biomedical Research Centre, Sumy State University, R-Korsakova Street, 40007 Sumy, Ukraine;
- Institute of Atomic Physics and Spectroscopy, University of Latvia, Jelgavas Iela 3, LV-1004 Riga, Latvia
| |
Collapse
|
11
|
Chen J, Xing Y, Bai X, Xue M, Shi Q, Li B. Strong Bioactive Glass-Based Hybrid Implants with Good Biomineralization Activity Used to Reduce Formation Duration and Improve Biomechanics of Bone Regeneration. Polymers (Basel) 2023; 15:3497. [PMID: 37688122 PMCID: PMC10489730 DOI: 10.3390/polym15173497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Developing bioactive implants with strong mechanical properties and biomineralization activity is critical in bone repair. In this work, modified cellulose nanofiber (mCNF)-reinforced bioactive glass (BG)-polycaprolactone (PCL) hybrids (mCNF-BP) with strong biomechanics and good apatite formation ability were reported. Incorporating mCNFs shortens the forming duration of the hybrid films and enhances the biomechanical performance and in vitro apatite-formation capability. The optimized biomechanical performance of the optimal hybrid materials is produced at a relatively high mCNF content (1.0 wt%), including a considerably higher modulus of elasticity (948.65 ± 74.06 MPa). In addition, the biomineralization activity of mCNF-BP hybrids is also tailored with the increase in the mCNF contents. The mCNF-BP with 1.5 wt% and 2.0 wt% mCNFs demonstrate the best biomineralization activity after immersing in simulated body fluid for 3 days. This study suggests that mCNFs are efficient bioactive additive to reinforce BG-based hybrids' mechanical properties and biomineralization activity.
Collapse
Affiliation(s)
- Jing Chen
- The Key Laboratory for Surface Engineering and Remanufacturing in Shaanxi Province, School of Chemical Engineering, Xi’an University, Xi’an 710065, China; (X.B.); (M.X.); (Q.S.); (B.L.)
| | - Yonglei Xing
- State Key Laboratory of High-Efficiency Utilization of Coal and Green Chemical Engineering, National Demonstration Center for Experimental Chemistry Education, School of Chemistry and Chemical Engineering, Ningxia University, Yinchuan 750021, China
| | - Xiaozhuan Bai
- The Key Laboratory for Surface Engineering and Remanufacturing in Shaanxi Province, School of Chemical Engineering, Xi’an University, Xi’an 710065, China; (X.B.); (M.X.); (Q.S.); (B.L.)
| | - Min Xue
- The Key Laboratory for Surface Engineering and Remanufacturing in Shaanxi Province, School of Chemical Engineering, Xi’an University, Xi’an 710065, China; (X.B.); (M.X.); (Q.S.); (B.L.)
| | - Qi Shi
- The Key Laboratory for Surface Engineering and Remanufacturing in Shaanxi Province, School of Chemical Engineering, Xi’an University, Xi’an 710065, China; (X.B.); (M.X.); (Q.S.); (B.L.)
| | - Beibei Li
- The Key Laboratory for Surface Engineering and Remanufacturing in Shaanxi Province, School of Chemical Engineering, Xi’an University, Xi’an 710065, China; (X.B.); (M.X.); (Q.S.); (B.L.)
| |
Collapse
|
12
|
Yazdanpanah Z, Sharma NK, Raquin A, Cooper DML, Chen X, Johnston JD. Printing tissue-engineered scaffolds made of polycaprolactone and nano-hydroxyapatite with mechanical properties appropriate for trabecular bone substitutes. Biomed Eng Online 2023; 22:73. [PMID: 37474951 PMCID: PMC10360269 DOI: 10.1186/s12938-023-01135-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Bone tissue engineering, based on three-dimensional (3D) printing technology, has emerged as a promising approach to treat bone defects using scaffolds. The objective of this study was to investigate the influence of porosity and internal structure on the mechanical properties of scaffolds. METHODS We fabricated composite scaffolds (which aimed to replicate trabecular bone) from polycaprolactone (PCL) reinforced with 30% (wt.) nano-hydroxyapatite (nHAp) by extrusion printing. Scaffolds with various porosities were designed and fabricated with and without an interlayer offset, termed as staggered and lattice structure, respectively. Mechanical compressive testing was performed to determine scaffold elastic modulus and yield strength. Linear regression was used to evaluate mechanical properties as a function of scaffold porosity. RESULTS Different relationships between mechanical properties and porosities were noted for the staggered and lattice structures. For elastic moduli, the two relationships intersected (porosity = 55%) such that the lattice structure exhibited higher moduli with porosity values greater than the intersection point; vice versa for the staggered structure. The lattice structure exhibited higher yield strength at all porosities. Mechanical testing results also indicated elastic moduli and yield strength properties comparable to trabecular bone (elastic moduli: 14-165 MPa; yield strength: 0.9-10 MPa). CONCLUSIONS Taken together, this study demonstrates that scaffolds printed from PCL/30% (wt.) nHAp with lattice and staggered structure offer promise for treating trabecular bone defects. This study identified the effect of porosity and internal structure on scaffold mechanical properties and provided suggestions for developing scaffolds with mechanical properties for substituting trabecular bone.
Collapse
Affiliation(s)
- Zahra Yazdanpanah
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada.
| | - Nitin Kumar Sharma
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Alice Raquin
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Institut Catholique Des Arts Et Métiers, 85000, La Roche-Sur-Yon, France
| | - David M L Cooper
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - James D Johnston
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
13
|
Grilli F, Albanesi E, Pelacho B, Prosper F, Decuzzi P, Di Mascolo D. Microstructured Polymeric Fabrics Modulating the Paracrine Activity of Adipose-Derived Stem Cells. Int J Mol Sci 2023; 24:10123. [PMID: 37373273 DOI: 10.3390/ijms241210123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The deposition of stem cells at sites of injury is a clinically relevant approach to facilitate tissue repair and angiogenesis. However, insufficient cell engraftment and survival require the engineering of novel scaffolds. Here, a regular network of microscopic poly(lactic-co-glycolic acid) (PLGA) filaments was investigated as a promising biodegradable scaffold for human Adipose-Derived Stem Cell (hADSC) tissue integration. Via soft lithography, three different microstructured fabrics were realized where 5 × 5 and 5 × 3 μm PLGA 'warp' and 'weft' filaments crossed perpendicularly with pitch distances of 5, 10 and 20 μm. After hADSC seeding, cell viability, actin cytoskeleton, spatial organization and the secretome were characterized and compared to conventional substrates, including collagen layers. On the PLGA fabric, hADSC re-assembled to form spheroidal-like structures, preserving cell viability and favoring a nonlinear actin organization. Moreover, the secretion of specific factors involved in angiogenesis, the remodeling of the extracellular matrix and stem cell homing was favored on the PLGA fabric as compared to that which occurred on conventional substrates. The paracrine activity of hADSC was microstructure-dependent, with 5 μm PLGA fabric enhancing the expression of factors involved in all three processes. Although more studies are needed, the proposed PLGA fabric would represent a promising alternative to conventional collagen substrates for stem cell implantation and angiogenesis induction.
Collapse
Affiliation(s)
- Federica Grilli
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genoa, 16145 Genoa, Italy
| | - Ennio Albanesi
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Beatriz Pelacho
- Laboratory of Regenerative Medicine, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Felipe Prosper
- Laboratory of Regenerative Medicine, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy
- Department of Electrical and Information Engineering, Politecnico di Bari, 70126 Bari, Italy
| |
Collapse
|
14
|
He Y, Liang L, Luo C, Zhang ZY, Huang J. Strategies for in situ tissue engineering of vascularized bone regeneration (Review). Biomed Rep 2023; 18:42. [PMID: 37325184 PMCID: PMC10265129 DOI: 10.3892/br.2023.1625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/03/2023] [Indexed: 06/17/2023] Open
Abstract
Numerous physiological processes occur following bone fracture, including inflammatory cell recruitment, vascularization, and callus formation and remodeling. In particular circumstances, such as critical bone defects or osteonecrosis, the regenerative microenvironment is compromised, rendering endogenous stem/progenitor cells incapable of fully manifesting their reparative potential. Consequently, external interventions, such as grafting or augmentation, are frequently necessary. In situ bone tissue engineering (iBTE) employs cell-free scaffolds that possess microenvironmental cues, which, upon implantation, redirect the behavior of endogenous stem/progenitor cells towards a pro-regenerative inflammatory response and reestablish angiogenesis-osteogenesis coupling. This process ultimately results in vascularized bone regeneration (VBR). In this context, a comprehensive review of the current techniques and modalities in VBR-targeted iBTE technology is provided.
Collapse
Affiliation(s)
- Yijun He
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Lin Liang
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| | - Cheng Luo
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| | - Zhi-Yong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Jiongfeng Huang
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| |
Collapse
|
15
|
Intravaia JT, Graham T, Kim HS, Nanda HS, Kumbar SG, Nukavarapu SP. Smart Orthopedic Biomaterials and Implants. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 25:100439. [PMID: 36642994 PMCID: PMC9835562 DOI: 10.1016/j.cobme.2022.100439] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Musculoskeletal injuries including bone defects continue to present a significant challenge in orthopedic surgery due to suboptimal healing. Bone reconstruction strategies focused on the use of biological grafts and bone graft substitutes in the form of biomaterials-based 3D structures in fracture repair. Recent advances in biomaterials science and engineering have resulted in the creation of intricate 3D bone-mimicking structures that are mechanically stable, biodegradable, and bioactive to support bone regeneration. Current efforts are focused on improving the biomaterial and implant physicochemical properties to promote interactions with the host tissue and osteogenesis. The "smart" biomaterials and their 3D structures are designed to actively interact with stem/progenitor cells and the extracellular matrix (ECM) to influence the local environment towards osteogenesis and de novo tissue formation. This article will summarize such smart biomaterials and the methodologies to apply either internal or external stimuli to control the tissue healing microenvironment. A particular emphasis is also made on the use of smart biomaterials and strategies to create functional bioactive implants for bone defect repair and regeneration.
Collapse
Affiliation(s)
| | - Trevon Graham
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Hyun S. Kim
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Himansu S. Nanda
- Terasaki Institute, University of California, Los Angeles, CA, USA
- Mechanical Engineering, IIITDM, Jabalpur, MP, India
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Syam P. Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
16
|
Inada Y, Takabatake K, Tsujigiwa H, Nakano K, Shan Q, Piao T, Chang A, Kawai H, Nagatsuka H. Novel Artificial Scaffold for Bone Marrow Regeneration: Honeycomb Tricalcium Phosphate. MATERIALS (BASEL, SWITZERLAND) 2023; 16:1393. [PMID: 36837023 PMCID: PMC9965701 DOI: 10.3390/ma16041393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/23/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Bone marrow is complex structure containing heterogenetic cells, making it difficult to regenerate using artificial scaffolds. In a previous study, we succeeded in developing honeycomb tricalcium phosphate (TCP), which is a cylindrical scaffold with a honeycomb arrangement of straight pores, and we demonstrated that TCP with 300 and 500 μm pore diameters (300TCP and 500TCP) induced bone marrow structure within the pores. In this study, we examined the optimal scaffold structure for bone marrow with homeostatic bone metabolism using honeycomb TCP. 300TCP and 500TCP were transplanted into rat muscle, and bone marrow formation was histologically assessed. Immunohistochemistry for CD45, CD34, Runt-related transcription factor 2 (Runx2), c-kit single staining, Runx2/N-cadherin, and c-kit/Tie-2 double staining was performed. The area of bone marrow structure, which includes CD45(+) round-shaped hematopoietic cells and CD34(+) sinusoidal vessels, was larger in 300TCP than in 500TCP. Additionally, Runx2(+) osteoblasts and c-kit(+) hematopoietic stem cells were observed on the surface of bone tissue formed within TCP. Among Runx2(+) osteoblasts, spindle-shaped N-cadherin(+) cells existed in association with c-kit(+)Tie-2(+) hematopoietic stem cells on the bone tissue formed within TCP, which formed a hematopoietic stem cell niche similar to as in vivo. Therefore, honeycomb TCP with 300 μm pore diameters may be an artificial scaffold with an optimal geometric structure as a scaffold for bone marrow formation.
Collapse
Affiliation(s)
- Yasunori Inada
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Kiyofumi Takabatake
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Hidetsugu Tsujigiwa
- Department of Life Science, Faculty of Science, Okayama University of Science, Okayama 700-0005, Japan
| | - Keisuke Nakano
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Qiusheng Shan
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Tianyan Piao
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Anqi Chang
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Hotaka Kawai
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Hitoshi Nagatsuka
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| |
Collapse
|
17
|
The effect of extracellular matrix remodeling on material-based strategies for bone regeneration: Review article. Tissue Cell 2022; 76:101748. [DOI: 10.1016/j.tice.2022.101748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 01/14/2022] [Accepted: 01/31/2022] [Indexed: 11/22/2022]
|
18
|
Ansari MAA, Golebiowska AA, Dash M, Kumar P, Jain PK, Nukavarapu SP, Ramakrishna S, Nanda HS. Engineering biomaterials to 3D-print scaffolds for bone regeneration: practical and theoretical consideration. Biomater Sci 2022; 10:2789-2816. [PMID: 35510605 DOI: 10.1039/d2bm00035k] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
There are more than 2 million bone grafting procedures performed annually in the US alone. Despite significant efforts, the repair of large segmental bone defects is a substantial clinical challenge which requires bone substitute materials or a bone graft. The available biomaterials lack the adequate mechanical strength to withstand the static and dynamic loads while maintaining sufficient porosity to facilitate cell in-growth and vascularization during bone tissue regeneration. A wide range of advanced biomaterials are being currently designed to mimic the physical as well as the chemical composition of a bone by forming polymer blends, polymer-ceramic and polymer-degradable metal composites. Transforming these novel biomaterials into porous and load-bearing structures via three-dimensional printing (3DP) has emerged as a popular manufacturing technique to develop engineered bone grafts. 3DP has been adopted as a versatile tool to design and develop bone grafts that satisfy porosity and mechanical requirements while having the ability to form grafts of varied shapes and sizes to meet the physiological requirements. In addition to providing surfaces for cell attachment and eventual bone formation, these bone grafts also have to provide physical support during the repair process. Hence, the mechanical competence of the 3D-printed scaffold plays a key role in the success of the implant. In this review, we present various recent strategies that have been utilized to design and develop robust biomaterials that can be deployed for 3D-printing bone substitutes. The article also reviews some of the practical, theoretical and biological considerations adopted in the 3D-structure design and development for bone tissue engineering.
Collapse
Affiliation(s)
- Mohammad Aftab Alam Ansari
- Biomedical Engineering and Technology Lab, Mechanical engineering discipline, PDPM Indian Institute of Information Technology, Design & Manufacturing Jabalpur, India.
- FFF Laboratory, Mechanical engineering discipline, PDPM Indian Institute of Information Technology, Design & Manufacturing Jabalpur, India.
- International Centre for Sustainable and Net Zero Technologies, PDPM-Indian Institute of Information Technology Design and Manufacturing (IIITDM) Jabalpur, Dumna Airport Road, Jabalpur-482005, MP, India
| | - Aleksandra A Golebiowska
- Biomedical Engineering, Materials Science & Engineering, and Orthopaedic Surgery, University of Connecticut, 260 Glenbrook Road, Unit 3247 Storrs, CT, 06269, USA
| | - Madhusmita Dash
- School of Minerals, Metallurgical and Materials Engineering, Indian Institute of Technology Bhubaneswar, Arugul, Khurdha 752050, Odisha, India
- International Centre for Sustainable and Net Zero Technologies, PDPM-Indian Institute of Information Technology Design and Manufacturing (IIITDM) Jabalpur, Dumna Airport Road, Jabalpur-482005, MP, India
| | - Prasoon Kumar
- Biodesign and Medical device laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Prashant Kumar Jain
- FFF Laboratory, Mechanical engineering discipline, PDPM Indian Institute of Information Technology, Design & Manufacturing Jabalpur, India.
| | - Syam P Nukavarapu
- Biomedical Engineering, Materials Science & Engineering, and Orthopaedic Surgery, University of Connecticut, 260 Glenbrook Road, Unit 3247 Storrs, CT, 06269, USA
| | - Seeram Ramakrishna
- Centre for Nanofibers and Nanotechnology, Department of Mechanical Engineering, National University of Singapore, Engineering Drive 3, Singapore 117587, Singapore
| | - Himansu Sekhar Nanda
- Biomedical Engineering and Technology Lab, Mechanical engineering discipline, PDPM Indian Institute of Information Technology, Design & Manufacturing Jabalpur, India.
- International Centre for Sustainable and Net Zero Technologies, PDPM-Indian Institute of Information Technology Design and Manufacturing (IIITDM) Jabalpur, Dumna Airport Road, Jabalpur-482005, MP, India
| |
Collapse
|
19
|
Golebiowska AA, Nukavarapu SP. Bio-inspired zonal-structured matrices for bone-cartilage interface engineering. Biofabrication 2022; 14:025016. [PMID: 35147514 DOI: 10.1088/1758-5090/ac5413] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/08/2022] [Indexed: 11/11/2022]
Abstract
Design and development of scaffold structures for osteochondral (OC) interface regeneration is a significant engineering challenge. Recent efforts are aimed at recapitulating the unique compositional and hierarchical structure of an OC interface. Conventional scaffold fabrication techniques often have limited design control and reproducibility, and the development of OC scaffolds with zonal hierarchy and structural integrity between zones is especially challenging. In this study, a series of multi-zonal and gradient structures were designed and fabricated using three-dimensional bioprinting. We developed OC scaffolds with bi-phasic and tri-phasic configurations to support the zonal structure of OC tissue, and gradient scaffold configurations to enable smooth transitions between the zones to more closely mimic a bone-cartilage interface. A biodegradable polymer, polylactic acid, was used for the fabrication of zonal/gradient scaffolds to provide mechanical strength and support OC function. The formation of the multi-zonal and gradient scaffolds was confirmed through scanning electron microscopy imaging and micro-computed tomography scanning. Precisely controlled hierarchy with tunable porosity along the scaffold length established the formation of the bio-inspired scaffolds with different zones/gradient structure. In addition, we also developed a novel bioprinting method to selectively introduce cells into desired scaffold zones of the zonal/gradient scaffolds via concurrent printing of a cell-laden hydrogel within the porous template. Live/dead staining of the cell-laden hydrogel introduced in the cartilage zone showed uniform cell distribution with high cell viability. Overall, our study developed bio-inspired scaffold structures with structural hierarchy and mechanical integrity for bone-cartilage interface engineering.
Collapse
Affiliation(s)
- Aleksandra A Golebiowska
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT-06269, United States of America
| | - Syam P Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT-06269, United States of America
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT-06269, United States of America
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT-06032, United States of America
| |
Collapse
|
20
|
García-Henández AL, Serrano-Bello J, Alvarez-Perez MA, Cifuentes-Mendiola E, González-Alva P. The immune system and its response to polymeric materials used for craniofacial regeneration. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2021.2021904] [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]
Affiliation(s)
- Ana Lilia García-Henández
- Division of Postgraduate Studies and Dental Research, Faculty of Dentistry, Tissue Bioengineering Laboratory, National Autonomous University of Mexico, Mexico City, Mexico
| | - Janeth Serrano-Bello
- Section of Oral Immunity and Bone Regulation, Laboratory of Dental Research, FES Iztacala National Autonomous University of Mexico, CDMX, State of Mexico, Mexico
| | - Marco Antonio Alvarez-Perez
- Section of Oral Immunity and Bone Regulation, Laboratory of Dental Research, FES Iztacala National Autonomous University of Mexico, CDMX, State of Mexico, Mexico
| | - Ernesto Cifuentes-Mendiola
- Division of Postgraduate Studies and Dental Research, Faculty of Dentistry, Tissue Bioengineering Laboratory, National Autonomous University of Mexico, Mexico City, Mexico
| | - Patricia González-Alva
- Section of Oral Immunity and Bone Regulation, Laboratory of Dental Research, FES Iztacala National Autonomous University of Mexico, CDMX, State of Mexico, Mexico
| |
Collapse
|
21
|
Polylactide, Processed by a Foaming Method Using Compressed Freon R134a, for Tissue Engineering. Polymers (Basel) 2021; 13:polym13203453. [PMID: 34685212 PMCID: PMC8539307 DOI: 10.3390/polym13203453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 11/17/2022] Open
Abstract
Fabricating polymeric scaffolds using cost-effective manufacturing processes is still challenging. Gas foaming techniques using supercritical carbon dioxide (scCO2) have attracted attention for producing synthetic polymer matrices; however, the high-pressure requirements are often a technological barrier for its widespread use. Compressed 1,1,1,2-tetrafluoroethane, known as Freon R134a, offers advantages over CO2 in manufacturing processes in terms of lower pressure and temperature conditions and the use of low-cost equipment. Here, we report for the first time the use of Freon R134a for generating porous polymer matrices, specifically polylactide (PLA). PLA scaffolds processed with Freon R134a exhibited larger pore sizes, and total porosity, and appropriate mechanical properties compared with those achieved by scCO2 processing. PLGA scaffolds processed with Freon R134a were highly porous and showed a relatively fragile structure. Human mesenchymal stem cells (MSCs) attached to PLA scaffolds processed with Freon R134a, and their metabolic activity increased during culturing. In addition, MSCs displayed spread morphology on the PLA scaffolds processed with Freon R134a, with a well-organized actin cytoskeleton and a dense matrix of fibronectin fibrils. Functionalization of Freon R134a-processed PLA scaffolds with protein nanoparticles, used as bioactive factors, enhanced the scaffolds' cytocompatibility. These findings indicate that gas foaming using compressed Freon R134a could represent a cost-effective and environmentally friendly fabrication technology to produce polymeric scaffolds for tissue engineering approaches.
Collapse
|
22
|
Pharmaceutical electrospinning and 3D printing scaffold design for bone regeneration. Adv Drug Deliv Rev 2021; 174:504-534. [PMID: 33991588 DOI: 10.1016/j.addr.2021.05.007] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Bone regenerative engineering provides a great platform for bone tissue regeneration covering cells, growth factors and other dynamic forces for fabricating scaffolds. Diversified biomaterials and their fabrication methods have emerged for fabricating patient specific bioactive scaffolds with controlled microstructures for bridging complex bone defects. The goal of this review is to summarize the points of scaffold design as well as applications for bone regeneration based on both electrospinning and 3D bioprinting. It first briefly introduces biological characteristics of bone regeneration and summarizes the applications of different types of material and the considerations for bone regeneration including polymers, ceramics, metals and composites. We then discuss electrospinning nanofibrous scaffold applied for the bone regenerative engineering with various properties, components and structures. Meanwhile, diverse design in the 3D bioprinting scaffolds for osteogenesis especially in the role of drug and bioactive factors delivery are assembled. Finally, we discuss challenges and future prospects in the development of electrospinning and 3D bioprinting for osteogenesis and prominent strategies and directions in future.
Collapse
|
23
|
Duarte MM, Ribeiro N, Silva IV, Dias JR, Alves NM, Oliveira AL. Fast decellularization process using supercritical carbon dioxide for trabecular bone. J Supercrit Fluids 2021. [DOI: 10.1016/j.supflu.2021.105194] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
24
|
Jeyakumar V, Amraish N, Niculescu-Morsza E, Bauer C, Pahr D, Nehrer S. Decellularized Cartilage Extracellular Matrix Incorporated Silk Fibroin Hybrid Scaffolds for Endochondral Ossification Mediated Bone Regeneration. Int J Mol Sci 2021; 22:ijms22084055. [PMID: 33919985 PMCID: PMC8071030 DOI: 10.3390/ijms22084055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/10/2021] [Accepted: 04/10/2021] [Indexed: 11/29/2022] Open
Abstract
Tissue engineering strategies promote bone regeneration for large bone defects by stimulating the osteogenesis route via intramembranous ossification in engineered grafts, which upon implantation are frequently constrained by insufficient integration and functional anastomosis of vasculature from the host tissue. In this study, we developed a hybrid biomaterial incorporating decellularized cartilage extracellular matrix (CD-ECM) as a template and silk fibroin (SF) as a carrier to assess the bone regeneration capacity of bone marrow-derived mesenchymal stem cells (hBMSC’s) via the endochondral ossification (ECO) route. hBMSC’s were primed two weeks for chondrogenesis, followed by six weeks for hypertrophy onto hybrid CD-ECM/SF or SF alone scaffolds and evaluated for the mineralized matrix formation in vitro. Calcium deposition biochemically determined increased significantly from 4-8 weeks in both SF and CD-ECM/SF constructs, and retention of sGAG’s were observed only in CD-ECM/SF constructs. SEM/EDX revealed calcium and phosphate crystal localization by hBMSC’s under all conditions. Compressive modulus reached a maximum of 40 KPa after eight weeks of hypertrophic induction. μCT scanning at eight weeks indicated a cloud of denser minerals in groups after hypertrophic induction in CD-ECM/SF constructs than SF constructs. Gene expression by RT-qPCR revealed that hBMSC’s expressed hypertrophic markers VEGF, COL10, RUNX2, but the absence of early hypertrophic marker ChM1 and later hypertrophic marker TSBS1 and the presence of osteogenic markers ALPL, IBSP, OSX under all conditions. Our data indicate a new method to prime hBMSC’S into the late hypertrophic stage in vitro in mechanically stable constructs for ECO-mediated bone tissue regeneration.
Collapse
Affiliation(s)
- Vivek Jeyakumar
- Center for Regenerative Medicine, Danube University Krems, 3500 Krems, Austria; (E.N.-M.); (C.B.); (S.N.)
- Correspondence:
| | - Nedaa Amraish
- Department of Anatomy and Biomechanics, Karl Landsteiner University for Health Sciences, 3500 Krems, Austria; (N.A.); (D.P.)
- Institute for Lightweight Design and Structural Biomechanics, Vienna University of Technology, 1060 Vienna, Austria
| | - Eugenia Niculescu-Morsza
- Center for Regenerative Medicine, Danube University Krems, 3500 Krems, Austria; (E.N.-M.); (C.B.); (S.N.)
| | - Christoph Bauer
- Center for Regenerative Medicine, Danube University Krems, 3500 Krems, Austria; (E.N.-M.); (C.B.); (S.N.)
| | - Dieter Pahr
- Department of Anatomy and Biomechanics, Karl Landsteiner University for Health Sciences, 3500 Krems, Austria; (N.A.); (D.P.)
- Institute for Lightweight Design and Structural Biomechanics, Vienna University of Technology, 1060 Vienna, Austria
| | - Stefan Nehrer
- Center for Regenerative Medicine, Danube University Krems, 3500 Krems, Austria; (E.N.-M.); (C.B.); (S.N.)
| |
Collapse
|
25
|
Zeinali R, del Valle LJ, Torras J, Puiggalí J. Recent Progress on Biodegradable Tissue Engineering Scaffolds Prepared by Thermally-Induced Phase Separation (TIPS). Int J Mol Sci 2021; 22:ijms22073504. [PMID: 33800709 PMCID: PMC8036748 DOI: 10.3390/ijms22073504] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/23/2022] Open
Abstract
Porous biodegradable scaffolds provide a physical substrate for cells allowing them to attach, proliferate and guide the formation of new tissues. A variety of techniques have been developed to fabricate tissue engineering (TE) scaffolds, among them the most relevant is the thermally-induced phase separation (TIPS). This technique has been widely used in recent years to fabricate three-dimensional (3D) TE scaffolds. Low production cost, simple experimental procedure and easy processability together with the capability to produce highly porous scaffolds with controllable architecture justify the popularity of TIPS. This paper provides a general overview of the TIPS methodology applied for the preparation of 3D porous TE scaffolds. The recent advances in the fabrication of porous scaffolds through this technique, in terms of technology and material selection, have been reviewed. In addition, how properties can be effectively modified to serve as ideal substrates for specific target cells has been specifically addressed. Additionally, examples are offered with respect to changes of TIPS procedure parameters, the combination of TIPS with other techniques and innovations in polymer or filler selection.
Collapse
Affiliation(s)
- Reza Zeinali
- Departament d’Enginyeria Química, Universitat Politècnica de Catalunya, Escola d’Enginyeria de Barcelona Est-EEBE, 08019 Barcelona, Spain; (L.J.d.V.); (J.T.)
- Correspondence: (R.Z.); (J.P.); Tel.: +34-93-401-1620 (R.Z.); +34-93-401-5649 (J.P.)
| | - Luis J. del Valle
- Departament d’Enginyeria Química, Universitat Politècnica de Catalunya, Escola d’Enginyeria de Barcelona Est-EEBE, 08019 Barcelona, Spain; (L.J.d.V.); (J.T.)
| | - Joan Torras
- Departament d’Enginyeria Química, Universitat Politècnica de Catalunya, Escola d’Enginyeria de Barcelona Est-EEBE, 08019 Barcelona, Spain; (L.J.d.V.); (J.T.)
| | - Jordi Puiggalí
- Departament d’Enginyeria Química, Universitat Politècnica de Catalunya, Escola d’Enginyeria de Barcelona Est-EEBE, 08019 Barcelona, Spain; (L.J.d.V.); (J.T.)
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, c/Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Correspondence: (R.Z.); (J.P.); Tel.: +34-93-401-1620 (R.Z.); +34-93-401-5649 (J.P.)
| |
Collapse
|
26
|
Pandya M, Saxon M, Bozanich J, Tillberg C, Luan X, Diekwisch TG. The Glycoprotein/Cytokine Erythropoietin Promotes Rapid Alveolar Ridge Regeneration In Vivo by Promoting New Bone Extracellular Matrix Deposition in Conjunction with Coupled Angiogenesis/Osteogenesis. Int J Mol Sci 2021; 22:2788. [PMID: 33801825 PMCID: PMC8002021 DOI: 10.3390/ijms22062788] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/20/2022] Open
Abstract
The loss of bone following tooth extraction poses a significant clinical problem for maxillofacial esthetics, function, and future implant placement. In the present study, the efficacy of an erythropoietin-impregnated collagen scaffold as an alveolar ridge augmentation material versus a conventional collagen scaffold and a BioOss inorganic bovine bone xenograft was examined. The collagen/Erythropoietin (EPO) scaffold exhibited significantly more rapid and complete osseous regeneration of the alveolar defect when compared to bone xenograft and the collagen membrane alone. The new EPO induced extracellular matrix was rich in Collagen I, Collagen III, Fibronectin (Fn) and E-cadherin, and featured significantly increased levels of the osteogenic transcription factors Runt-related transcription factor 2 (Runx2) and Osterix (Osx). Histomorphometric evaluation revealed a significant two-fold increase in the number of capillaries between the EPO and the BioOss group. Moreover, there was a highly significant 3.5-fold higher level of vascular endothelial growth factor (VEGF) in the collagen/EPO-treated group compared to controls. The significant effect of EPO on VEGF, FN, and RUNX2 upregulation was confirmed in vitro, and VEGF pathway analysis using VEGF inhibitors confirmed that EPO modulated extracellular matrix protein expression through VEGF even in the absence of blood vessels. Together, these data demonstrate the effectiveness of an EPO-impregnated collagen scaffold for bone regeneration as it induces rapid matrix production and osseoinduction adjacent to new capillaries via VEGF.
Collapse
Affiliation(s)
- Mirali Pandya
- Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA; (M.P.); (C.T.); (X.L.)
- Department of Periodontics, Texas A&M College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA; (M.S.); (J.B.)
| | - Matthew Saxon
- Department of Periodontics, Texas A&M College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA; (M.S.); (J.B.)
| | - John Bozanich
- Department of Periodontics, Texas A&M College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA; (M.S.); (J.B.)
| | - Connie Tillberg
- Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA; (M.P.); (C.T.); (X.L.)
| | - Xianghong Luan
- Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA; (M.P.); (C.T.); (X.L.)
- Department of Periodontics, Texas A&M College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA; (M.S.); (J.B.)
| | - Thomas G.H. Diekwisch
- Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA; (M.P.); (C.T.); (X.L.)
- Department of Periodontics, Texas A&M College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA; (M.S.); (J.B.)
| |
Collapse
|
27
|
Dorcemus DL, Kim HS, Nukavarapu SP. Gradient scaffold with spatial growth factor profile for osteochondral interface engineering. Biomed Mater 2020; 16. [PMID: 33291092 DOI: 10.1088/1748-605x/abd1ba] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/08/2020] [Indexed: 11/11/2022]
Abstract
Osteochondral (OC) matrix design poses a significant engineering challenge due to the complexity involved with bone-cartilage interfaces. To better facilitate the regeneration of OC tissue, we developed and evaluated a biodegradable matrix with uniquely arranged bone and cartilage supporting phases: a poly(lactic-co-glycolic) acid (PLGA) template structure with a porosity gradient along its longitudinal axis uniquely integrated with hyaluronic acid hydrogel. Micro-CT scanning and imaging confirmed the formation of an inverse gradient matrix. Hydroxyapatite was added to the PLGA template which was then plasma-treated to increase hydrophilicity and growth factor affinity. An osteogenic growth factor (bone morphogenetic protein 2; BMP-2) was loaded onto the template scaffold via adsorption, while a chondrogenic growth factor (transforming growth factor beta 1; TGF-β1) was incorporated into the hydrogel phase. Confocal microscopy of the growth factor loaded matrix confirmed the spatial distribution of the two growth factors, with chondrogenic factor confined to the cartilaginous portion and osteogenic factor present throughout the scaffold. We observed spatial differentiation of human mesenchymal stem cells (hMSCs) into cartilage and bone cells in the scaffolds in vitro: cartilaginous regions were marked by increased glycosaminoglycan production, and osteogenesis was seen throughout the graft by alizarin red staining. In a dose-dependent study of BMP-2, hMSC pellet cultures with TGF-β1 and BMP-2 showed synergistic effects on chondrogenesis. These results indicate that development of an inverse gradient matrix can spatially distribute two different growth factors to facilitate chondrogenesis and osteogenesis along different portions of a scaffold, which are key steps needed for formation of an osteochondral interface.
Collapse
Affiliation(s)
- Deborah Leonie Dorcemus
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, Connecticut, 06269, UNITED STATES
| | - Hyun Sung Kim
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, Connecticut, 06269, UNITED STATES
| | - Syam Prasad Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, Connecticut, 06269, UNITED STATES
| |
Collapse
|
28
|
Daneshmandi L, Shah S, Jafari T, Bhattacharjee M, Momah D, Saveh-Shemshaki N, Lo KWH, Laurencin CT. Emergence of the Stem Cell Secretome in Regenerative Engineering. Trends Biotechnol 2020; 38:1373-1384. [PMID: 32622558 PMCID: PMC7666064 DOI: 10.1016/j.tibtech.2020.04.013] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
The secretome is defined as the set of molecules and biological factors that are secreted by cells into the extracellular space. In the past decade, secretome-based therapies have emerged as a promising approach to overcome the limitations associated with cell-based therapies for tissue and organ regeneration. Considering the growing number of recent publications related to secretome-based therapies, this review takes a step-by-step engineering approach to evaluate the role of the stem cell secretome in regenerative engineering. We discuss the functional benefits of the secretome, the techniques used to engineer the secretome and tailor its therapeutic effects, and the delivery systems and strategies that have been developed to use the secretome for tissue regeneration.
Collapse
Affiliation(s)
- Leila Daneshmandi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Shiv Shah
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA; Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Tahereh Jafari
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Maumita Bhattacharjee
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Deandra Momah
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
| | - Nikoo Saveh-Shemshaki
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Kevin W-H Lo
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Institute of Materials Science, University of Connecticut, Storrs, CT 06269
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA; Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA; Institute of Materials Science, University of Connecticut, Storrs, CT 06269; Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Medicine, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
29
|
Distinct Osteogenic Potentials of BMP-2 and FGF-2 in Extramedullary and Medullary Microenvironments. Int J Mol Sci 2020; 21:ijms21217967. [PMID: 33120952 PMCID: PMC7662681 DOI: 10.3390/ijms21217967] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/24/2020] [Accepted: 10/24/2020] [Indexed: 12/14/2022] Open
Abstract
Bone morphogenetic protein-2 (BMP-2) and fibroblast growth factor-2 (FGF-2) have been regarded as the major cytokines promoting bone formation, however, several studies have reported unexpected results with failure of bone formation or bone resorption of these growth factors. In this study, BMP-2 and FGF-2 adsorbed into atellocollagen sponges were transplanted into bone defects in the bone marrow-scarce calvaria (extramedullary environment) and bone marrow-abundant femur (medullary environment) for analysis of their in vivo effects not only on osteoblasts, osteoclasts but also on bone marrow cells. The results showed that BMP-2 induced high bone formation in the bone marrow-scarce calvaria, but induced bone resorption in the bone marrow-abundant femurs. On the other hand, FGF-2 showed opposite effects compared to those of BMP-2. Analysis of cellular dynamics revealed numerous osteoblasts and osteoclasts present in the newly-formed bone induced by BMP-2 in calvaria, but none were seen in either control or FGF-2-transplanted groups. On the other hand, in the femur, numerous osteoclasts were observed in the vicinity of the BMP-2 pellet, while a great number of osteoblasts were seen near the FGF-2 pellets or in the control group. Of note, FCM analysis showed that both BMP-2 and FGF-2 administrated in the femur did not significantly affect the hematopoietic cell population, indicating a relatively safe application of the two growth factors. Together, these results indicate that BMP-2 could be suitable for application in extramedullary bone regeneration, whereas FGF-2 could be suitable for application in medullary bone regeneration.
Collapse
|
30
|
Mikael PE, Golebiowska AA, Kumbar SG, Nukavarapu SP. Evaluation of Autologously Derived Biomaterials and Stem Cells for Bone Tissue Engineering. Tissue Eng Part A 2020; 26:1052-1063. [PMID: 32375566 PMCID: PMC7580602 DOI: 10.1089/ten.tea.2020.0011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/16/2020] [Indexed: 11/12/2022] Open
Abstract
Despite progress, clinical translation of tissue engineering (TE) products/technologies is limited. A significant effort is underway to develop biomaterials and cells through a minimally modified process for clinical translation of TE products. Recently, bone marrow aspirate (BMA) was identified as an autologous source of cells for TE applications and is currently being tested in clinical therapies, but the isolation methods need improvement to avoid potential for contamination and increase progenitor cell yield. To address these issues, we reproducibly processed human peripheral blood (PB) and BMA to develop autologously derived biomaterials and cells. We demonstrated PB-derived biomaterial/gel cross-linking and fibrin gel formation with varied gelation times as well as biocompatibility through support of human bone marrow-derived stem cell survival and growth in vitro. Next, we established a plastic culture-free process that concentrates and increases the yield of CD146+/CD271+ early mesenchymal progenitor cells in BMA (concentrated BMA [cBMA]). cBMA exhibited increased colony formation and multipotency (including chondrogenic differentiation) in vitro compared with standard BMA. PB-derived gels encapsulated with cBMA also demonstrated increased cell proliferation and enhanced mineralization when assessed for bone TE in vitro. This strategy can potentially be developed for use in any tissue regeneration application; however, bone regeneration was used as a test bed for this study.
Collapse
Affiliation(s)
- Paiyz E. Mikael
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
| | | | - Sangamesh G. Kumbar
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, Connecticut, USA
| | - Syam P. Nukavarapu
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, Connecticut, USA
| |
Collapse
|
31
|
Dhandapani R, Krishnan PD, Zennifer A, Kannan V, Manigandan A, Arul MR, Jaiswal D, Subramanian A, Kumbar SG, Sethuraman S. Additive manufacturing of biodegradable porous orthopaedic screw. Bioact Mater 2020; 5:458-467. [PMID: 32280835 PMCID: PMC7139166 DOI: 10.1016/j.bioactmat.2020.03.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/11/2020] [Accepted: 03/18/2020] [Indexed: 01/04/2023] Open
Abstract
Advent of additive manufacturing in biomedical field has nurtured fabrication of complex, customizable and reproducible orthopaedic implants. Layer-by-layer deposition of biodegradable polymer employed in development of porous orthopaedic screws promises gradual dissolution and complete metabolic resorption thereby overcoming the limitations of conventional metallic screws. In the present study, screws with different pore sizes (916 × 918 μm to 254 × 146 μm) were 3D printed at 200 μm layer height by varying printing parameters such as print speed, fill density and travel speed to augment the bone ingrowth. Micro-CT analysis and scanning electron micrographs of screws with 45% fill density confirmed porous interconnections (40.1%) and optimal pore size (259 × 207 × 200 μm) without compromising the mechanical strength (24.58 ± 1.36 MPa). Due to the open pore structure, the 3D printed screws showed increased weight gain due to the deposition of calcium when incubated in simulated body fluid. Osteoblast-like cells attached on screw and infiltrated into the pores over 14 days of in vitro culture. Further, the screws also supported greater human mesenchymal stem cell adhesion, proliferation and mineralized matrix synthesis over a period of 21 days in vitro culture as compared to non-porous screws. These porous screws showed significantly increased vascularization in a rat subcutaneous implantation as compared to control screws. Porous screws produced by additive manufacturing may promote better osteointegration due to enhanced mineralization and vascularization.
Collapse
Affiliation(s)
- Ramya Dhandapani
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, 613401, India
| | - Priya Dharshini Krishnan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, 613401, India
| | - Allen Zennifer
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, 613401, India
| | - Vishal Kannan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, 613401, India
| | - Amrutha Manigandan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, 613401, India
| | - Michael R. Arul
- Department of Orthopaedics, UConn Health, Farmington, CT, 06030, USA
| | - Devina Jaiswal
- Department of Orthopaedics, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, Western New England University, Springfield, MA, 01119, USA
| | - Anuradha Subramanian
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, 613401, India
| | | | - Swaminathan Sethuraman
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, 613401, India
| |
Collapse
|
32
|
Robust phenotypic maintenance of limb cells during heterogeneous culture in a physiologically relevant polymeric-based constructed graft system. Sci Rep 2020; 10:11739. [PMID: 32678185 PMCID: PMC7367281 DOI: 10.1038/s41598-020-68658-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/29/2020] [Indexed: 11/08/2022] Open
Abstract
A major challenge during the simultaneous regeneration of multiple tissues is the ability to maintain the phenotypic characteristics of distinct cell populations on one construct, especially in the presence of different exogenous soluble cues such as growth factors. Therefore, in this study, we questioned whether phenotypic maintenance over a distinct population of cells can be achieved by providing biomimetic structural cues relevant to each cell phenotype into the construct's design and controlling the presentation of growth factors in a region-specific manner. To address this question, we developed a polymeric-based constructed graft system (CGS) as a physiologically relevant model that consists of three combined regions with distinct microstructures and growth factor types. Regions A and B of the CGS exhibited similar microstructures to the skin and soft tissues and contained rhPDGF-BB and rhIGF-I, while region C exhibited a similar microstructure to the bone tissue and contained rhBMP-2. Primary rat skin fibroblasts, soft tissue fibroblasts, and osteoblasts were then cultured on regions A, B, and C of the CGS, respectively and their phenotypic characteristics were evaluated in this heterogenous environment. In the absence of growth factors, we found that the structural cues presented in every region played a key role in maintaining the region-specific cell functions and heterogeneity during a heterogeneous culture. In the presence of growth factors, we found that spatially localizing the growth factors at their respective regions resulted in enhanced region-specific cell functions and maintained region-specific cell heterogeneity compared to supplementation, which resulted in a significant reduction of cell growth and loss of phenotype. Our data suggest that providing biomimetic structural cues relevant to each cell phenotype and controlling the presentation of growth factors play a crucial role in ensuring heterogeneity maintenance of distinct cell populations during a heterogeneous culture. The presented CGS herein provides a reliable platform for investigating different cells responses to heterogeneous culture in a physiologically relevant microenvironment. In addition, the model provides a unique platform for evaluating the feasibility and efficacy of different approaches for simultaneously delivering multiple growth factors or molecules from a single construct to achieve enhanced cell response while maintaining cellular heterogeneity during a heterogenous culture.
Collapse
|
33
|
Shams M, Karimi M, Heydari M, Salimi A. Nanocomposite scaffolds composed of Apacite (apatite-calcite) nanostructures, poly (ε-caprolactone) and poly (2-hydroxyethylmethacrylate): The effect of nanostructures on physico-mechanical properties and osteogenic differentiation of human bone marrow mesenchymal stem cells in vitro. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 117:111271. [PMID: 32919635 DOI: 10.1016/j.msec.2020.111271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 01/05/2023]
Abstract
Nanocomposite scaffolds were fabricated from poly (ε-caprolactone) (PCL), Poly (2-hydroxyethylmethacrylate) (PHEMA), and Apacite (Apatite-calcite) nanostructures (15 and 25 wt%). The nanoscale structure, physical and chemical properties, mechanical properties, hydrophilic behavior, degradability and osteogenic properties of the fabricated scaffolds were evaluated. The results showed that the mechanical strength, degradation, wetting ability, and mechanical strength of PCL-PHEMA scaffolds significantly increases upon inclusion of Apacite nanoparticles up to 25 wt%. Accordingly, the best mechanical values (E ~ 7.109 MPa and σ ~ 0.414 MPa) and highest degradability (32% within 96 h) were recorded for PCL-PHEMA scaffolds containing 25 wt% of Apacite. Furthermore, highest porosity and roughness were observed in the PCL-PHEMA/25% Apacite as a result of the Apacite nanoparticles inclusion. There was no cytotoxicity recorded for the fabricated scaffolds based on the results obtained from MTT assay and acridine orange staining. Alkaline phosphatase activity, calcium content quantification, Van Kossa staining, FESEM and real time PCR tests confirmed the biomineralization, and the differentiation potential of the nanocomposite scaffolds. Overall, the 3D structure, optimum porosity and balanced dissolution rate of PCL-PHEMA/25% Apacite providing a balanced microenvironment resulted in improved cell adhesion, cell behavior, and replication, as well as osteogenic induction of human bone-marrow-derived mesenchymal stem cells (hBM-MSCs).
Collapse
Affiliation(s)
- Mehdi Shams
- Department of Nanotechnology and Advanced Materials, Materials and Energy Research Center, Karaj, Iran
| | - Mohammad Karimi
- Department of Nanotechnology and Advanced Materials, Materials and Energy Research Center, Karaj, Iran
| | - Masoomeh Heydari
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Bracey DN, Cignetti NE, Jinnah AH, Stone AV, Gyr BM, Whitlock PW, Scott AT. Bone xenotransplantation: A review of the history, orthopedic clinical literature, and a single‐center case series. Xenotransplantation 2020; 27:e12600. [DOI: 10.1111/xen.12600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/22/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Daniel N. Bracey
- Department of Orthopaedic Surgery Wake Forest School of Medicine Winston‐Salem NC USA
| | - Natalie E. Cignetti
- Department of Orthopaedic Surgery Wake Forest School of Medicine Winston‐Salem NC USA
| | - Alexander H. Jinnah
- Department of Orthopaedic Surgery Wake Forest School of Medicine Winston‐Salem NC USA
| | - Austin V. Stone
- Department of Orthopaedic Surgery and Sports Medicine University of Kentucky Lexington KY USA
| | - Bettina M. Gyr
- Department of Orthopedic Surgery and Sports Medicine Children’s Hospital of the King’s Daughters Norfolk VA USA
| | - Patrick W. Whitlock
- Division of Orthopaedic Surgery Cincinnati Children’s Hospital Medical Center Cincinnati OH USA
| | - Aaron T. Scott
- Department of Orthopaedic Surgery Wake Forest School of Medicine Winston‐Salem NC USA
| |
Collapse
|
35
|
Hayashi K, Munar ML, Ishikawa K. Effects of macropore size in carbonate apatite honeycomb scaffolds on bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110848. [PMID: 32279778 DOI: 10.1016/j.msec.2020.110848] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/13/2020] [Accepted: 03/12/2020] [Indexed: 01/23/2023]
Abstract
The pore architecture of scaffolds is a critical factor for angiogenesis and bone regeneration. Although the effects of scaffold macropore size have been investigated, most scaffolds feature macropores with poor uniformity and interconnectivity, and other parameters (e.g., microporosity, chemical composition, and strut thickness) differ among scaffolds. To clarify the threshold of effective macropore size, we fabricated honeycomb scaffolds (HCSs) with distinct macropore (i.e., channel) sizes (~100, ~200, and ~300 μm). The HCSs were composed of AB-type carbonate apatite with ~8.5% carbonate ions, i.e., the same composition as human bone mineral. Their honeycomb architecture displayed uniformly sized and orderly arranged channels with extremely high interconnectivity, and all the HCSs displayed ~100-μm-thick struts and 0.06 cm3 g-1 of micropore volume. The compressive strengths of HCSs with ~100-, ~200-, and ~300-μm channels were higher than those of reported scaffolds, and decreased with increasing channel size: 62 ± 6, 55 ± 9, and 43 ± 8 MPa, respectively. At four weeks after implantation in rabbit femur bone defects, new bone and blood vessels were formed in all the channels of these HCSs. Notably, the ~300-μm channels were extensively occupied by new bone. We demonstrated that high interconnectivity and uniformity of channels can decrease the threshold of effective macropore size, enabling the scaffolds to maintain high mechanical properties and osteogenic ability and serve as implants for weight-bearing areas.
Collapse
Affiliation(s)
- Koichiro Hayashi
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.
| | - Melvin L Munar
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kunio Ishikawa
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
36
|
Lee DJ, Kwon J, Kim YI, Wang X, Wu TJ, Lee YT, Kim S, Miguez P, Ko CC. Effect of pore size in bone regeneration using polydopamine-laced hydroxyapatite collagen calcium silicate scaffolds fabricated by 3D mould printing technology. Orthod Craniofac Res 2019; 22 Suppl 1:127-133. [PMID: 31074145 DOI: 10.1111/ocr.12261] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 12/04/2018] [Indexed: 01/14/2023]
Abstract
OBJECTIVE The pore size of the scaffold is a critical factor in repairing large bone defect. Here, we investigated the potential of bone regeneration using novel nanocomposite polydopamine-laced hydroxyapatite collagen calcium silicate (HCCS-PDA) scaffolds with two different pore sizes, 250 and 500 μm. SAMPLES/SETTING A total of 12 male Sprague-Dawley rats were implanted with HCCS-PDA scaffold with pore size of either 250 or 500 μm into surgically created critical-sized defect (CSD). METHODS HCCS-PDA scaffolds were fabricated using mould printing technique. The effect of pore size on mechanical strength of the scaffolds was assessed by compression testing. After seeding with rat mesenchymal stem cells (rMSCs), the scaffolds were implanted, and new bone formation was evaluated using microCT and histomorphometric analysis after 8 weeks. RESULTS MicroCT and histology analysis demonstrated restricted peripheral new bone formation in either dural or periosteal side and limited new bone formation in the 250 μm pore scaffold. Conversely, the 500-μm pore scaffold showed more penetration of new bone into the scaffold and greater bone regeneration in the rat CSD. CONCLUSION Based on our results, which demonstrated improved new bone formation in 500 μm pores scaffold, we can conclude that effective scaffold pore size that induces osteointegration and bone regeneration is around 500 μm for HCCS-PDA nanocomposite scaffold.
Collapse
Affiliation(s)
- Dong Joon Lee
- Oral and Craniofacial Health Sciences Research, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina
| | - Jane Kwon
- Oral and Craniofacial Health Sciences Research, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina
| | - Yong-Il Kim
- Oral and Craniofacial Health Sciences Research, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina.,Department of Orthodontics, School of Dentistry, Pusan National University, Yangsan, Republic of Korea
| | - Xiaoyu Wang
- Oral and Craniofacial Health Sciences Research, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina
| | - Te-Ju Wu
- Oral and Craniofacial Health Sciences Research, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina.,Department of Orthodontics, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yan-Ting Lee
- Oral and Craniofacial Health Sciences Research, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina
| | - Steven Kim
- Oral and Craniofacial Health Sciences Research, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina
| | - Patricia Miguez
- Oral and Craniofacial Health Sciences Research, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina
| | - Ching-Chang Ko
- Oral and Craniofacial Health Sciences Research, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina.,Department of Orthodontics, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
37
|
Rosenberg M, Shilo D, Galperin L, Capucha T, Tarabieh K, Rachmiel A, Segal E. Bone Morphogenic Protein 2-Loaded Porous Silicon Carriers for Osteoinductive Implants. Pharmaceutics 2019; 11:E602. [PMID: 31726775 PMCID: PMC6920899 DOI: 10.3390/pharmaceutics11110602] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/19/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are probably the most important growth factors in bone formation and healing. However, the utilization of BMPs in clinical applications is mainly limited due to the protein poor solubility at physiological pH, rapid clearance and relatively short biological half-life. Herein, we develop degradable porous silicon (PSi)-based carriers for sustained delivery of BMP-2. Two different loading approaches are examined, physical adsorption and covalent conjugation, and their effect on the protein loading and release rate is thoroughly studied. The entrapment of the protein within the PSi nanostructures preserved its bioactivity for inducing osteogenic differentiation of rabbit bone marrow mesenchymal stems cells (BM-MSCs). BM-MSCs cultured with the BMP-2 loaded PSi carriers exhibit a relatively high alkaline phosphatase (ALP) activity. We also demonstrate that exposure of MSCs to empty PSi (no protein) carriers generates some extent of differentiation due to the ability of the carrier's degradation products to induce osteoblast differentiation. Finally, we demonstrate the integration of these promising BMP-2 carriers within a 3D-printed patient-specific implant, constructed of poly(caprolactone) (PCL), as a potential bone graft for critical size bone defects.
Collapse
Affiliation(s)
- Michal Rosenberg
- Department of Biotechnology and Food Engineering, Technion—Israel Institute of Technology, Haifa 3200003, Israel; (M.R.); (L.G.)
| | - Dekel Shilo
- Department of Oral and Maxillofacial Surgery, Rambam Health Care Campus, Haifa 3109601, Israel; (D.S.); (T.C.); (K.T.); (A.R.)
- Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3109601, Israel
| | - Leonid Galperin
- Department of Biotechnology and Food Engineering, Technion—Israel Institute of Technology, Haifa 3200003, Israel; (M.R.); (L.G.)
| | - Tal Capucha
- Department of Oral and Maxillofacial Surgery, Rambam Health Care Campus, Haifa 3109601, Israel; (D.S.); (T.C.); (K.T.); (A.R.)
| | - Karim Tarabieh
- Department of Oral and Maxillofacial Surgery, Rambam Health Care Campus, Haifa 3109601, Israel; (D.S.); (T.C.); (K.T.); (A.R.)
| | - Adi Rachmiel
- Department of Oral and Maxillofacial Surgery, Rambam Health Care Campus, Haifa 3109601, Israel; (D.S.); (T.C.); (K.T.); (A.R.)
- Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3109601, Israel
| | - Ester Segal
- Department of Biotechnology and Food Engineering, Technion—Israel Institute of Technology, Haifa 3200003, Israel; (M.R.); (L.G.)
- Russell Berrie Nanotechnology Institute, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
38
|
Celie KB, Toyoda Y, Dong X, Morrison KA, Zhang P, Asanbe O, Jin JL, Hooper RC, Zanotelli MR, Kaymakcalan O, Bender RJ, Spector JA. Microstructured hydrogel scaffolds containing differential density interfaces promote rapid cellular invasion and vascularization. Acta Biomater 2019; 91:144-158. [PMID: 31004845 DOI: 10.1016/j.actbio.2019.04.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Insufficient vascularization of currently available clinical biomaterials has limited their application to optimal wound beds. We designed a hydrogel scaffold with a unique internal microstructure of differential collagen densities to induce cellular invasion and neovascularization. METHODS Microsphere scaffolds (MSS) were fabricated by encasing 1% (w/v) type 1 collagen microspheres 50-150 μm in diameter in 0.3% collagen bulk. 1% and 0.3% monophase collagen scaffolds and Integra® disks served as controls. Mechanical characterization as well as in vitro and in vivo invasion assays were performed. Cell number and depth of invasion were analyzed using Imaris™. Cell identity was assessed immunohistochemically. RESULTS In vitro, MSS exhibited significantly greater average depth of cellular invasion than Integra® and monophase collagen controls. MSS also demonstrated significantly higher cell counts than controls. In vivo, MSS revealed significantly more cellular invasion spanning the entire scaffold depth at 14 days than Integra®. CD31+ expressing luminal structures suggestive of neovasculature were seen within MSS at 7 days and were more prevalent after 14 days. Multiphoton microscopy of MSS demonstrated erythrocytes within luminal structures after 14 days. CONCLUSION By harnessing simple architectural cues to induce cellular migration, MSS holds great potential for clinical translation as the next generation dermal replacement product. STATEMENT OF SIGNIFICANCE Large skin wounds require tissue engineered dermal substitutes in order to promote healing. Currently available dermal replacement products do not always adequately incorporate into the body, especially in complex wounds, due to poor neovascularization. In this paper, we present a hydrogel with an innovative microarchitecture that is composed of dense type I collagen microspheres suspended in a less-dense collagen bulk. We show that cell invasion into the scaffold is driven solely by mechanical cues inherent within this differential density interface, and that this induces robust vascular cell invasion both in vitro and in a rodent model. Our hydrogel performs favorably compared to the current clinical gold standard, Integra®. We believe this hydrogel scaffold may be the first of the next generation of dermal replacement products.
Collapse
Affiliation(s)
- Karel-Bart Celie
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Yoshiko Toyoda
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Xue Dong
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Kerry A Morrison
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Peipei Zhang
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Ope Asanbe
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Julia L Jin
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Rachel C Hooper
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 121A Weill Hall, Ithaca, NY 14853, United States
| | - Omer Kaymakcalan
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Ryan J Bender
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Jason A Spector
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 121A Weill Hall, Ithaca, NY 14853, United States.
| |
Collapse
|
39
|
Mikael PE, Golebiowska AA, Xin X, Rowe DW, Nukavarapu SP. Evaluation of an Engineered Hybrid Matrix for Bone Regeneration via Endochondral Ossification. Ann Biomed Eng 2019; 48:992-1005. [PMID: 31037444 DOI: 10.1007/s10439-019-02279-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/24/2019] [Indexed: 12/28/2022]
Abstract
Despite its regenerative ability, long and segmental bone defect repair remains a significant orthopedic challenge. Conventional tissue engineering efforts induce bone formation through intramembranous ossification (IO) which limits vascular formation and leads to poor bone regeneration. To overcome this challenge, a novel hybrid matrix comprised of a load-bearing polymer template and a gel phase is designed and assessed for bone regeneration. Our previous studies developed a synthetic ECM, hyaluronan (HA)-fibrin (FB), that is able to mimic cartilage-mediated bone formation in vitro. In this study, the well-characterized HA-FB hydrogel is combined with a biodegradable polymer template to form a hybrid matrix. In vitro evaluation of the matrix showed cartilage template formation, cell recruitment and recruited cell osteogenesis, essential stages in endochondral ossification. A transgenic reporter-mouse critical-defect model was used to evaluate the bone healing potential of the hybrid matrix in vivo. The results demonstrated host cell recruitment into the hybrid matrix that led to new bone formation and subsequent remodeling of the mineralization. Overall, the study developed and evaluated a novel load-bearing graft system for bone regeneration via endochondral ossification.
Collapse
Affiliation(s)
- Paiyz E Mikael
- Department of Materials Science, & Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Aleksandra A Golebiowska
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
| | - Xiaonan Xin
- Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health, Farmington, CT, 06032, USA
| | - David W Rowe
- Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Syam P Nukavarapu
- Department of Materials Science, & Engineering, University of Connecticut, Storrs, CT, 06269, USA. .,Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA.
| |
Collapse
|
40
|
Gandolfi MG, Zamparini F, Degli Esposti M, Chiellini F, Fava F, Fabbri P, Taddei P, Prati C. Highly porous polycaprolactone scaffolds doped with calcium silicate and dicalcium phosphate dihydrate designed for bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 102:341-361. [PMID: 31147007 DOI: 10.1016/j.msec.2019.04.040] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 01/31/2019] [Accepted: 04/12/2019] [Indexed: 01/24/2023]
Abstract
Polycaprolactone (PCL), dicalcium phosphate dihydrate (DCPD) and/or calcium silicates (CaSi) have been used to prepare highly porous scaffolds by thermally induced phase separation technique (TIPS). Three experimental mineral-doped formulations were prepared (PCL-10CaSi, PCL-5CaSi-5DCPD, PCL-10CaSi-10DCPD); pure PCL scaffolds constituted the control group. Scaffolds were tested for their chemical-physical and biological properties, namely thermal properties by differential scanning calorimetry (DSC), mechanical properties by quasi-static parallel-plates compression testing, porosity by a standard water-absorption method calcium release, alkalinizing activity, surface microchemistry and micromorphology by Environmental Scanning electronic Microscopy (ESEM), apatite-forming ability in Hank Balanced Saline Solution (HBSS) by Energy Dispersive X-ray Spectroscopy (EDX) and micro-Raman, and direct contact cytotoxicity. All mineral-doped scaffolds released calcium and alkalinized the soaking medium, which may favor a good biological (osteogenic) response. ESEM surface micromorphology analyses after soaking in HBSS revealed: pure PCL, PCL-10CaSi and PCL-10CaSi-10DCPD kept similar surface porosity percentages but different pore shape modifications. PCL-5CaSi-5DCPD revealed a significant surface porosity increase despite calcium phosphates nucleation (p < 0.05). Micro-Raman spectroscopy detected the formation of a B-type carbonated apatite (Ap) layer on the surface of PCL-10CaSi-10DCPD aged for 28 days in HBSS; a similar phase (but of lower thickness) formed also on PCL-5CaSi-5DCPD and PCL; the deposit formed on PCL-10CaSi was mainly composed of calcite. All PCL showed bulk open porosity higher than 94%; however, no relevant brittleness was observed in the materials, which retained the possibility to be handled without collapsing. The thermo-mechanical properties showed that the reinforcing and nucleating action of the inorganic fillers CaSi and DCPD improved viscoelastic properties of the scaffolds, as confirmed by the increased value of storage modulus and the slight increase in the crystallization temperature for all the biomaterials. A detrimental effect on the mechanical properties was observed in samples with the highest amount of inorganic particles (PCL-10CaSi-10DCPD). All the scaffolds showed absence of toxicity, in particular PCL-10CaSi-10DCPD. The designed scaffolds are biointeractive (release biologically relevant ions), nucleate apatite, possess high surface and internal open porosity and can be colonized by cells, creating a bone forming osteoblastic microenvironment and appearing interesting materials for bone regeneration purposes.
Collapse
Affiliation(s)
- Maria Giovanna Gandolfi
- Laboratory of Biomaterials and Oral Pathology, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| | - Fausto Zamparini
- Laboratory of Biomaterials and Oral Pathology, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy; Endodontic Clinical Section, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Micaela Degli Esposti
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Bologna, Italy
| | - Federica Chiellini
- BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Fabio Fava
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Bologna, Italy
| | - Paola Fabbri
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Bologna, Italy
| | - Paola Taddei
- Biochemistry Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carlo Prati
- Endodontic Clinical Section, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
41
|
Hatton J, Davis GR, Mourad AHI, Cherupurakal N, Hill RG, Mohsin S. Fabrication of Porous Bone Scaffolds Using Alginate and Bioactive Glass. J Funct Biomater 2019; 10:jfb10010015. [PMID: 30836701 PMCID: PMC6462929 DOI: 10.3390/jfb10010015] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/10/2019] [Accepted: 01/31/2019] [Indexed: 12/27/2022] Open
Abstract
Porous composite scaffold using an alginate and bioactive glass ICIE16M was synthesized by a simple freeze-drying technique. The scaffold was characterized using compression testing, Fourier-transform infrared spectroscopy (FTIR), differential scanning calorimetry (DSC), X-ray diffraction (XRD), X-ray microtomography (XMT) and scanning electron microscopy (SEM). The bioactivity of the scaffold was evaluated by its ability to form apatite on its surface in simulated body fluid (SBF). The data collected showed evidence that the novel material produced had an appropriate pore size for osteoconduction, with an average pore size of 110 µm and maximum pore size of 309 µm. Statistical analysis confirmed that the glass filler significantly (P < 0.05) increased the collapse yield of the scaffolds compared with pure alginate scaffolds. The ICIE16M glass had an amorphous structure, favorable for bioactivity.
Collapse
Affiliation(s)
- Jonathan Hatton
- Dental Physical Sciences Unit, Institute of Dentistry, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 4NS, UK.
| | - Graham Roy Davis
- Dental Physical Sciences Unit, Institute of Dentistry, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 4NS, UK.
| | - Abdel-Hamid I Mourad
- Department of Mechanical Engineering, College of Engineering, UAEU Al Ain, 15551, UAE.
| | | | - Robert G Hill
- Dental Physical Sciences Unit, Institute of Dentistry, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 4NS, UK.
| | - Sahar Mohsin
- Dental Physical Sciences Unit, Institute of Dentistry, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 4NS, UK.
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, 17666, UAE.
| |
Collapse
|
42
|
Li X, Zhang Y, Zhong K, Lu S, Chen Y. [Study on the bone regeneration induced by advanced-platelet-rich fibrin and β- tricalcium phosphate composite]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:177-184. [PMID: 30739411 PMCID: PMC8337601 DOI: 10.7507/1002-1892.201807002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 12/24/2018] [Indexed: 02/05/2023]
Abstract
Objective To explore the osteogenesis effect of advanced-platelet-rich fibrin (A-PRF) and β-tricalcium phosphate (β-TCP) composite. Methods Thirty-two healthy female New Zealand rabbits were randomly selected. A-PRF was prepared by collecting blood from middle auricular artery. Rabbits were randomly divided into 6 groups: groups A, B, C, D, and E (6 rabbits in each group) and group F (2 rabbits). Bone defects (6 mm in diameter, 8 mm in depth) were drilled into femur condyle of each rabbit's both back legs. Then A-PRF and β-TCP composites manufactured by different proportion were planted into bone defects of group A (1∶1), group B (2∶1), group C (4∶1), group D (1∶2) and group E (1∶4) ( V/ V). The bone defect was not repaired in group F. The specimens were collected at 8 and at 12 weeks after operation. Then gross observation, X-ray examination, Micro-CT examination, and biomechanical test were performed. The bone volume/total volume (BV/TV), trabecular number (Tb.N), trabecular thickness (Tb.Th), and trabecular spacing (Tb.Sp), compressive strength, and modulus of elasticity were calculated. Results The gross observation and X-ray examination showed that the osteogenesis effect at 12 weeks was better than that at 8 weeks. At the same time point, the repair of bone defect and the formation of new bone in group B were better than those in other groups. Micro-CT examination showed that the trabeculae of new bone in group B were the most and the trabeculae arranged closely at 8 and 12 weeks. Besides there were significant differences in BV/TV, Tb.N, and Tb.Sp between group B and the other groups ( P<0.05). There were significant differences in Tb.N and Tb.Th in group B, BV/TV and Tb.Sp in group C, Tb.Sp in group D between 8 weeks and 12 weeks ( P<0.05). Biomechanical tests showed that the compression strength and elastic modulus of group B were the highest, and the compression strength and elastic modulus of group C were the lowest at 8 and at 12 weeks, showing significant differences ( P<0.05). There were significant differences in compression strength and elastic modulus of each group between 8 weeks and 12 weeks ( P<0.05). Conclusion The A-PRF and β-TCP composite can repair femoral condylar defects in rabbits, and the osteogenesis is better in proportion of 2∶1.
Collapse
Affiliation(s)
- Xuemei Li
- School of Stomatology, Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Yushi Zhang
- School of Stomatology, Southwest Medical University, Luzhou Sichuan, 646000, P.R.China;Department of Stomatology, Chengdu Hospital of Sichuan Petroleum Administration, Chengdu Sichuan, 610051, P.R.China
| | - Ke Zhong
- School of Stomatology, Southwest Medical University, Luzhou Sichuan, 646000,
| | - Shuai Lu
- Department of Stomatology, Authority Hospital of Chengdu Military Region, Chengdu Sichuan, 610031, P.R.China
| | - Yue Chen
- Department of Nuclear Medicine & Sichuan Key Laboratory of Nuclear Medicine and Molecular Imaging, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| |
Collapse
|
43
|
Ho-Shui-Ling A, Bolander J, Rustom LE, Johnson AW, Luyten FP, Picart C. Bone regeneration strategies: Engineered scaffolds, bioactive molecules and stem cells current stage and future perspectives. Biomaterials 2018; 180:143-162. [PMID: 30036727 PMCID: PMC6710094 DOI: 10.1016/j.biomaterials.2018.07.017] [Citation(s) in RCA: 562] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/06/2018] [Accepted: 07/10/2018] [Indexed: 12/25/2022]
Abstract
Bone fractures are the most common traumatic injuries in humans. The repair of bone fractures is a regenerative process that recapitulates many of the biological events of embryonic skeletal development. Most of the time it leads to successful healing and the recovery of the damaged bone. Unfortunately, about 5-10% of fractures will lead to delayed healing or non-union, more so in the case of co-morbidities such as diabetes. In this article, we review the different strategies to heal bone defects using synthetic bone graft substitutes, biologically active substances and stem cells. The majority of currently available reviews focus on strategies that are still at the early stages of development and use mostly in vitro experiments with cell lines or stem cells. Here, we focus on what is already implemented in the clinics, what is currently in clinical trials, and what has been tested in animal models. Treatment approaches can be classified in three major categories: i) synthetic bone graft substitutes (BGS) whose architecture and surface can be optimized; ii) BGS combined with bioactive molecules such as growth factors, peptides or small molecules targeting bone precursor cells, bone formation and metabolism; iii) cell-based strategies with progenitor cells combined or not with active molecules that can be injected or seeded on BGS for improved delivery. We review the major types of adult stromal cells (bone marrow, adipose and periosteum derived) that have been used and compare their properties. Finally, we discuss the remaining challenges that need to be addressed to significantly improve the healing of bone defects.
Collapse
Affiliation(s)
- Antalya Ho-Shui-Ling
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France; CNRS, LMGP, 3 Parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Johanna Bolander
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium
| | - Laurence E Rustom
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 1304 West Springfield Avenue, Urbana, IL 61801, USA
| | - Amy Wagoner Johnson
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, 1206 West Green Street, Urbana, IL 61081, USA; Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA
| | - Frank P Luyten
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium.
| | - Catherine Picart
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France; CNRS, LMGP, 3 Parvis Louis Néel, 38031 Grenoble Cedex 01, France.
| |
Collapse
|
44
|
Bracey DN, Seyler TM, Jinnah AH, Lively MO, Willey JS, Smith TL, Van Dyke ME, Whitlock PW. A Decellularized Porcine Xenograft-Derived Bone Scaffold for Clinical Use as a Bone Graft Substitute: A Critical Evaluation of Processing and Structure. J Funct Biomater 2018; 9:jfb9030045. [PMID: 30002336 PMCID: PMC6164666 DOI: 10.3390/jfb9030045] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Bone grafts are used in approximately one half of all musculoskeletal surgeries. Autograft bone is the historic gold standard but is limited in supply and its harvest imparts significant morbidity to the patient. Alternative sources of bone graft include allografts, synthetics and, less commonly, xenografts which are taken from animal species. Xenografts are available in unlimited supply from healthy animal donors with controlled biology, avoiding the risk of human disease transmission, and may satisfy current demand for bone graft products. METHODS In the current study, cancellous bone was harvested from porcine femurs and subjected to a novel decellularization protocol to derive a bone scaffold. RESULTS The scaffold was devoid of donor cellular material on histology and DNA sampling (p < 0.01). Microarchitectural properties important for osteoconductive potential were preserved after decellularization as shown by high resolution imaging modalities. Proteomics data demonstrated similar profiles when comparing the porcine bone scaffold against commercially available human demineralized bone matrix approved for clinical use. CONCLUSION We are unaware of any porcine-derived bone graft products currently used in orthopaedic surgery practice. Results from the current study suggest that porcine-derived bone scaffolds warrant further consideration to serve as a potential bone graft substitute.
Collapse
Affiliation(s)
- Daniel N Bracey
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | - Thorsten M Seyler
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Alexander H Jinnah
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | - Mark O Lively
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | - Jeffrey S Willey
- Department of Radiation Oncology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27157, USA.
| | - Thomas L Smith
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | - Mark E Van Dyke
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA.
| | - Patrick W Whitlock
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
45
|
Vashisth P, Bellare JR. Development of hybrid scaffold with biomimetic 3D architecture for bone regeneration. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1325-1336. [DOI: 10.1016/j.nano.2018.03.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/16/2018] [Accepted: 03/29/2018] [Indexed: 01/27/2023]
|
46
|
Abstract
Craniofacial bones, separate from the appendicular skeleton, bear a significant amount of strain and stress generated from mastication-related muscles. Current research on the regeneration of craniofacial bone focuses on the reestablishment of an elaborate vascular network. In this review, current challenges and efforts particularly in advances of scaffold properties and techniques for vascularization remodeling in craniofacial bone tissue engineering will be discussed. A microenvironment of ischemia and hypoxia in the biomaterial core drives propagation and reorganization of endothelial progenitor cells (EPCs) to assemble into a primitive microvascular framework. Co-culture strategies and delivery of vasculogenic molecules enhance EPCs' differentiation and stimulate the host regenerative response to promote vessel sprouting and strength. To optimize structural and vascular integration, well-designed microstructures of scaffolds are biologically considered. Proper porous structures, matrix stiffness, and surface morphology of scaffolds have a profound influence on cell behaviors and thus affect revascularization. In addition, advanced techniques facilitating angiogenesis and vaculogenesis have also been discussed. Oxygen delivery biomaterials, scaffold-free cell sheet techniques, and arteriovenous loop-induced axial vascularization strategies bring us new understanding and powerful strategies to manage revascularization of large craniofacial bone defects. Although promising histological results have been achieved, the efficient perfusion and functionalization of newly formed vessels are still challenging.
Collapse
Affiliation(s)
- T Tian
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - T Zhang
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Lin
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X Cai
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
47
|
Xu TO, Kim HS, Stahl T, Nukavarapu SP. Self-neutralizing PLGA/magnesium composites as novel biomaterials for tissue engineering. Biomed Mater 2018; 13:035013. [PMID: 29362293 PMCID: PMC5884090 DOI: 10.1088/1748-605x/aaaa29] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Controlling acidic degradation of biodegradable polyesters remains a major clinical challenge. This work presents a simple and effective strategy of developing polyester composites with biodegradable magnesium metal or alloys. PLGA samples with compositions of 1, 3, 5, and 10 wt% magnesium were produced using a simple solvent-casting method, which resulted in composite films with near uniform Mg metal/alloy particle dispersion. Degradation study of the composite films showed that all compositions higher than 1 wt% magnesium were able to extend the duration of degradation, and buffer acidic pH resulting from PLGA degradation. PLGA composite with 5 wt% of magnesium showed near-neutral degradation pattern under sink conditions. Magnesium addition also showed improved mechanical characteristics in terms of the tensile modulus. In vitro experiments conducted by seeding PLGA composites with MC3T3-E1 pre-osteoblasts demonstrated increased ALP expression and cellular mineralization. The established new biodegradable polymer-metal system provides a useful biomaterial platform with a wide range of applications in biomedical device development and scaffold-based tissue engineering.
Collapse
Affiliation(s)
- Thomas O Xu
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington CT, United States of America. These authors contributed equally
| | | | | | | |
Collapse
|
48
|
Aravamudhan A, Ramos DM, Nip J, Kalajzic I, Kumbar SG. Micro-Nanostructures of Cellulose-Collagen for Critical Sized Bone Defect Healing. Macromol Biosci 2018; 18:10.1002/mabi.201700263. [PMID: 29178402 PMCID: PMC5835266 DOI: 10.1002/mabi.201700263] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/15/2017] [Indexed: 01/12/2023]
Abstract
Bone tissue engineering strategies utilize biodegradable polymeric matrices alone or in combination with cells and factors to provide mechanical support to bone, while promoting cell proliferation, differentiation, and tissue ingrowth. The performance of mechanically competent, micro-nanostructured polymeric matrices, in combination with bone marrow stromal cells (BMSCs), is evaluated in a critical sized bone defect. Cellulose acetate (CA) is used to fabricate a porous microstructured matrix. Type I collagen is then allowed to self-assemble on these microstructures to create a natural polymer-based, micro-nanostructured matrix (CAc). Poly (lactic-co-glycolic acid) matrices with identical microstructures serve as controls. Significantly higher number of implanted host cells are distributed in the natural polymer based micro-nanostructures with greater bone density and more uniform cell distribution. Additionally, a twofold increase in collagen content is observed with natural polymer based scaffolds. This study establishes the benefits of natural polymer derived micro-nanostructures in combination with donor derived BMSCs to repair and regenerate critical sized bone defects. Natural polymer based materials with mechanically competent micro-nanostructures may serve as an alternative material platform for bone regeneration.
Collapse
Affiliation(s)
- Aja Aravamudhan
- Skeletal Cranial Biology, UConn Health, Farmington, CT-06030, US
| | - Daisy M. Ramos
- Materials Science and Engineering, University of Connecticut, Storrs, CT-06269, US
| | - Jonathan Nip
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT-06269, US
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, Uconn Health, Farmington, CT-06030, US
| | - Sangamesh G. Kumbar
- Skeletal Cranial Biology, UConn Health, Farmington, CT-06030, US
- Materials Science and Engineering, University of Connecticut, Storrs, CT-06269, US
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT-06269, US
- Department of Orthopaedics, UConn Health, Farmington, CT-06030, US
| |
Collapse
|
49
|
Pereira DR, Reis RL, Oliveira JM. Layered Scaffolds for Osteochondral Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1058:193-218. [DOI: 10.1007/978-3-319-76711-6_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
50
|
Nanoparticles-Based Systems for Osteochondral Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:209-217. [DOI: 10.1007/978-3-319-76735-2_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|