1
|
Raftery RM, Gonzalez Vazquez AG, Walsh DP, Chen G, Laiva AL, Keogh MB, O'Brien FJ. Mobilizing Endogenous Progenitor Cells Using pSDF1α-Activated Scaffolds Accelerates Angiogenesis and Bone Repair in Critical-Sized Bone Defects. Adv Healthc Mater 2024; 13:e2401031. [PMID: 38850118 DOI: 10.1002/adhm.202401031] [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: 03/19/2024] [Revised: 06/05/2024] [Indexed: 06/09/2024]
Abstract
Mobilizing endogenous progenitor cells to repair damaged tissue in situ has the potential to revolutionize the field of regenerative medicine, while the early establishment of a vascular network will ensure survival of newly generated tissue. In this study, a gene-activated scaffold containing a stromal derived factor 1α plasmid (pSDF1α), a pro-angiogenic gene that is also thought to be involved in the recruitment of mesenchymal stromal cells (MSCs) to sites of injury is described. It is shown that over-expression of SDF1α protein enhanced MSC recruitment and induced vessel-like structure formation by endothelial cells in vitro. When implanted subcutaneously, transcriptomic analysis reveals that endogenous MSCs are recruited and significant angiogenesis is stimulated. Just 1-week after implantation into a calvarial critical-sized bone defect, pSDF1α-activated scaffolds are recruited MSCs and rapidly activate angiogenic and osteogenic programs, upregulating Runx2, Dlx5, and Sp7. At the same time-point, pVEGF-activated scaffolds are recruited a variety of cell types, activating endochondral ossification. The early response induced by both scaffolds leads to complete bridging of the critical-sized bone defects within 4-weeks. The versatile cell-free gene-activated scaffold described in this study is capable of harnessing and enhancing the body's own regenerative capacity and has immense potential in a myriad of applications.
Collapse
Affiliation(s)
- Rosanne M Raftery
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
- iEd Hub and Department of Anatomy and Neuroscience, College of Medicine and Health, University College Cork, Cork, T12 CY82, Ireland
| | - Arlyng G Gonzalez Vazquez
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
| | - David P Walsh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
- Translational Research in Nanomedical Devices, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Ashang L Laiva
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Tisse Engineering Research Group, Royal College of Surgeons in Ireland - Medical University of Bahrain, Adliya, Bahrain
| | - Michael B Keogh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Tisse Engineering Research Group, Royal College of Surgeons in Ireland - Medical University of Bahrain, Adliya, Bahrain
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
| |
Collapse
|
2
|
Hauser PV, Zhao L, Chang HM, Yanagawa N, Hamon M. In Vivo Vascularization Chamber for the Implantation of Embryonic Kidneys. Tissue Eng Part C Methods 2024; 30:63-72. [PMID: 38062758 DOI: 10.1089/ten.tec.2023.0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Abstract
A major obstacle to the implantation of ex vivo engineered tissues is the incorporation of functional vascular supply to support the growth of new tissue and to minimize ischemic injury. Existing prevascularization systems, such as arteriovenous (AV) loop-based systems, require microsurgery, limiting their use to larger animals. We aimed to develop an implantable device that can be prevascularized to enable vascularization of tissues in small rodents, and test its application on the vascularization of embryonic kidneys. Implanting the chamber between the abdominal aorta and the inferior vena cava, we detected endothelial cells and vascular networks after 48 h of implantation. Loading the chamber with collagen I (C), Matrigel (M), or Matrigel + vascular endothelial growth factor) (MV) had a strong influence on vascularization speed: Chambers loaded with C took 7 days to vascularize, 4 days for chambers with M, and 2 days for chambers with MV. Implantation of E12.5 mouse embryonic kidneys into prevascularized chambers (C, MV) was followed with significant growth and ureteric branching over 22 days. In contrast, the growth of kidneys in non-prevascularized chambers was stunted. We concluded that our prevascularized chamber is a valuable tool for vascularizing implanted tissues and tissue-engineered constructs. Further optimization will be necessary to control the directional growth of vascular endothelial cells within the chamber and the vascularization grade. Impact Statement Vascularization of engineered tissue, or organoids, constructs is a major hurdle in tissue engineering. Failure of vascularization is associated with prolonged ischemia time and potential tissue damage due to hypoxic effects. The method presented, demonstrates the use of a novel chamber that allows rapid vascularization of native and engineered tissues. We hope that this technology helps to stimulate research in the field of tissue vascularization and enables researchers to generate larger engineered vascularized tissues.
Collapse
Affiliation(s)
- Peter Viktor Hauser
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Lifu Zhao
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
| | - Hsiao-Min Chang
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Norimoto Yanagawa
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Morgan Hamon
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| |
Collapse
|
3
|
Xu J, Shen J, Sun Y, Wu T, Sun Y, Chai Y, Kang Q, Rui B, Li G. In vivo prevascularization strategy enhances neovascularization of β-tricalcium phosphate scaffolds in bone regeneration. J Orthop Translat 2022; 37:143-151. [PMID: 36313532 PMCID: PMC9582585 DOI: 10.1016/j.jot.2022.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 08/12/2022] [Accepted: 09/01/2022] [Indexed: 12/01/2022] Open
Abstract
Background Neovascularization is critical for bone regeneration. Numerous studies have explored prevascularization preimplant strategies, ranging from calcium phosphate cement (CPC) scaffolds to co-culturing CPCs with stem cells. The aim of the present study was to evaluate an alternative in vivo prevascularization approach, using preimplant-prepared macroporous beta-tricalcium phosphate (β-TCP) scaffolds and subsequent transplantation in bone defect model. Methods The morphology of β-TCPs was characterized by scanning electron microscopy. After 3 weeks of prevascularization within a muscle pouch at the lateral size of rat tibia, we transplanted prevascularized macroporous β-TCPs in segmental tibia defects, using blank β-TCPs as a control. Extent of neovascularization was determined by angiography and immunohistochemical (IHC) evaluations. Tibia samples were collected at different time points for biomechanical, radiological, and histological analyses. RT-PCR and western blotting were used to evaluate angio- and osteo-specific markers. Results With macroporous β-TCPs, we documented more vascular and supporting tissue invasion in the macroporous β-TCPs with prior in vivo prevascularization. Radiography, biomechanical, IHC, and histological analyses revealed considerably more vascularity and bone consolidation in β-TCP scaffolds that had undergone the prevascularization step compared to the blank β-TCP scaffolds. Moreover, the prevascularization treatment remarkably upregulated mRNA and protein expression of BMP2 and vascular endothelial growth factor (VEGF) during bone regeneration. Conclusion This novel in vivo prevascularization strategy successfully accelerated vascular formation to bone regeneration. Our findings indicate that prevascularized tissue-engineered bone grafts have promising potential in clinical applications. The translational potential of this article This study indicates a novel in vivo prevascularization strategy for growing vasculature on β-TCP scaffolds to be used for repair of large segmental bone defects, might serve as a promising tissue-engineered bone grafts in the future.
Collapse
Affiliation(s)
- Jia Xu
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Junjie Shen
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - YunChu Sun
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tianyi Wu
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yuxin Sun
- Department of Orthopaedics and Traumatology, Bao-An District People's Hospital, Shenzhen, PR China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qinglin Kang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Biyu Rui
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Corresponding author. Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, PR China.
| | - Gang Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Corresponding author.
| |
Collapse
|
4
|
Novel structural designs of 3D-printed osteogenic graft for rapid angiogenesis. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00212-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
5
|
Amini Z, Lari R. A systematic review of decellularized allograft and xenograft–derived scaffolds in bone tissue regeneration. Tissue Cell 2021; 69:101494. [DOI: 10.1016/j.tice.2021.101494] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 01/09/2021] [Accepted: 01/10/2021] [Indexed: 12/26/2022]
|
6
|
The Application of a Bone Marrow Mesenchymal Stem Cell Membrane in the Vascularization of a Decellularized Tracheal Scaffold. Stem Cells Int 2021; 2021:6624265. [PMID: 33747094 PMCID: PMC7960062 DOI: 10.1155/2021/6624265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/27/2021] [Accepted: 02/14/2021] [Indexed: 12/18/2022] Open
Abstract
Airway stenosis is a common problem in the neonatal intensive care unit (NICU) and pediatric intensive care unit (PICU). A tissue-engineered trachea is a new therapeutic method and a research hotspot. Successful vascularization is the key to the application of a tissue-engineered trachea. However, successful vascularization studies lack a complete description. In this study, it was assumed that rabbit bone marrow mesenchymal stem cells were obtained and induced by ascorbic acid to detect the tissue structure, ultrastructure, and gene expression of the extracellular matrix. A vascular endothelial cell culture medium was added in vitro to induce the vascularization of the stem cell sheet (SCS), and the immunohistochemistry and gene expression of vascular endothelial cell markers were detected. At the same time, vascular growth-related factors were added and detected during SCS construction. After the SCS and decellularized tracheal (DT) were constructed, a tetrandrine allograft was performed to observe its vascularization potential. We established the architecture and identified rabbit bone marrow mesenchymal stem cell membranes by 14 days of ascorbic acid, studied the role of a vascularized membrane in inducing bone marrow mesenchymal stem cells by in vitro ascorbic acid, and assessed the role of combining the stem cell membranes and noncellular tracheal scaffolds in vivo. Fourteen experiments confirmed that cell membranes promote angiogenesis at gene level. The results of 21-day in vitro experiments showed that the composite tissue-engineered trachea had strong angiogenesis. In vivo experiments show that a composite tissue-engineered trachea has strong potential for angiogenesis. It promotes the understanding of diseases of airway stenosis and tissue-engineered tracheal regeneration in newborns and small infants.
Collapse
|
7
|
Amler AK, Dinkelborg PH, Schlauch D, Spinnen J, Stich S, Lauster R, Sittinger M, Nahles S, Heiland M, Kloke L, Rendenbach C, Beck-Broichsitter B, Dehne T. Comparison of the Translational Potential of Human Mesenchymal Progenitor Cells from Different Bone Entities for Autologous 3D Bioprinted Bone Grafts. Int J Mol Sci 2021; 22:E796. [PMID: 33466904 PMCID: PMC7830021 DOI: 10.3390/ijms22020796] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/28/2020] [Accepted: 01/11/2021] [Indexed: 02/08/2023] Open
Abstract
Reconstruction of segmental bone defects by autologous bone grafting is still the standard of care but presents challenges including anatomical availability and potential donor site morbidity. The process of 3D bioprinting, the application of 3D printing for direct fabrication of living tissue, opens new possibilities for highly personalized tissue implants, making it an appealing alternative to autologous bone grafts. One of the most crucial hurdles for the clinical application of 3D bioprinting is the choice of a suitable cell source, which should be minimally invasive, with high osteogenic potential, with fast, easy expansion. In this study, mesenchymal progenitor cells were isolated from clinically relevant human bone biopsy sites (explant cultures from alveolar bone, iliac crest and fibula; bone marrow aspirates; and periosteal bone shaving from the mastoid) and 3D bioprinted using projection-based stereolithography. Printed constructs were cultivated for 28 days and analyzed regarding their osteogenic potential by assessing viability, mineralization, and gene expression. While viability levels of all cell sources were comparable over the course of the cultivation, cells obtained by periosteal bone shaving showed higher mineralization of the print matrix, with gene expression data suggesting advanced osteogenic differentiation. These results indicate that periosteum-derived cells represent a highly promising cell source for translational bioprinting of bone tissue given their superior osteogenic potential as well as their minimally invasive obtainability.
Collapse
Affiliation(s)
- Anna-Klara Amler
- Department of Medical Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.-K.A.); (D.S.); (R.L.)
- Cellbricks GmbH, 13355 Berlin, Germany;
| | - Patrick H. Dinkelborg
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| | - Domenic Schlauch
- Department of Medical Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.-K.A.); (D.S.); (R.L.)
- Cellbricks GmbH, 13355 Berlin, Germany;
| | - Jacob Spinnen
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| | - Stefan Stich
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| | - Roland Lauster
- Department of Medical Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.-K.A.); (D.S.); (R.L.)
| | - Michael Sittinger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| | - Susanne Nahles
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
| | - Max Heiland
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
| | | | - Carsten Rendenbach
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
| | - Benedicta Beck-Broichsitter
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Oral and Maxillofacial Surgery, and Berlin Institute of Health, 13353 Berlin, Germany; (S.N.); (M.H.); (C.R.); (B.B.-B.)
| | - Tilo Dehne
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Department of Rheumatology, and Berlin Institute of Health, 10117 Berlin, Germany; (J.S.); (S.S.); (M.S.); (T.D.)
| |
Collapse
|
8
|
Taguchi T, Lopez MJ. An overview of de novo bone generation in animal models. J Orthop Res 2021; 39:7-21. [PMID: 32910496 PMCID: PMC7820991 DOI: 10.1002/jor.24852] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 02/04/2023]
Abstract
Some of the earliest success in de novo tissue generation was in bone tissue, and advances, facilitated by the use of endogenous and exogenous progenitor cells, continue unabated. The concept of one health promotes shared discoveries among medical disciplines to overcome health challenges that afflict numerous species. Carefully selected animal models are vital to development and translation of targeted therapies that improve the health and well-being of humans and animals alike. While inherent differences among species limit direct translation of scientific knowledge between them, rapid progress in ex vivo and in vivo de novo tissue generation is propelling revolutionary innovation to reality among all musculoskeletal specialties. This review contains a comparison of bone deposition among species and descriptions of animal models of bone restoration designed to replicate a multitude of bone injuries and pathology, including impaired osteogenic capacity.
Collapse
Affiliation(s)
- Takashi Taguchi
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary MedicineLouisiana State UniversityBaton RougeLouisianaUSA
| | - Mandi J. Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary MedicineLouisiana State UniversityBaton RougeLouisianaUSA
| |
Collapse
|
9
|
Midha S, Jain KG, Bhaskar N, Kaur A, Rawat S, Giri S, Basu B, Mohanty S. Tissue-specific mesenchymal stem cell-dependent osteogenesis in highly porous chitosan-based bone analogs. Stem Cells Transl Med 2020; 10:303-319. [PMID: 33049125 PMCID: PMC7848378 DOI: 10.1002/sctm.19-0385] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/05/2020] [Accepted: 03/10/2020] [Indexed: 12/19/2022] Open
Abstract
Among conventional fabrication techniques, freeze‐drying process has widely been investigated for polymeric implants. However, the understanding of the stem cell progenitor‐dependent cell functionality modulation and quantitative analysis of early osseointegration of highly porous scaffolds have not been explored. Here, we developed a novel, highly porous, multimaterial composite, chitosan/hydroxyapatite/polycaprolactone (CHT/HA/PCL). The in vitro studies have been performed using mesenchymal stem cells (MSCs) from three tissue sources: human bone marrow‐derived MSCs (BM‐MSCs), adipose‐derived MSCs (AD‐MSCs), and Wharton's jelly‐derived MSCs (WJ‐MSCs). Although cell attachment and metabolic activity [3‐4,5‐dimethylthiazol‐2yl‐(2,5 diphenyl‐2H‐tetrazoliumbromide) assay] were ore enhanced in WJ‐MSC‐laden CHT/HA/PCL composites, scanning electron microscopy, real‐time gene expression (alkaline phosphatase [ALP], collagen type I [Col I], osteocalcin [OCN], and bone morphogenetic protein 4 [BMP‐4]), and immunostaining (COL I, β‐CATENIN, OCN, and SCLEROSTIN [SOST]) demonstrated pronounced osteogenesis with terminal differentiation on BM‐MSC‐laden CHT/HA/PCL composites only. The enhanced cell functionality on CHT/HA/PCL composites was explained in terms of interplay among the surface properties and the optimal source of MSCs. In addition, osteogenesis in rat tibial model over 6 weeks confirmed a better ratio of bone volume to the total volume for BM‐MSC‐laden composites over scaffold‐only and defect‐only groups. The clinically conformant combination of 3D porous architecture with pore sizes varying in the range of 20 to 200 μm together with controlled in vitro degradation and early osseointegration establish the potential of CHT/HA/PCL composite as a potential cancellous bone analog.
Collapse
Affiliation(s)
- Swati Midha
- Stem Cell Facility (Department of Biotechnology-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Krishan G Jain
- Stem Cell Facility (Department of Biotechnology-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Nitu Bhaskar
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore, India
| | - Amtoj Kaur
- Stem Cell Facility (Department of Biotechnology-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Sonali Rawat
- Stem Cell Facility (Department of Biotechnology-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Shibashish Giri
- Department of Cell Techniques and Applied Stem Cell Biology, Centre for Biotechnology and Biomedicine, Medical faculty, University of Leipzig, Leipzig, Germany.,Department of Plastic Surgery and Hand Surgery, University Hospital Rechts der Isar, Technische Universität München, Munich, Germany
| | - Bikramjit Basu
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore, India
| | - Sujata Mohanty
- Stem Cell Facility (Department of Biotechnology-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
10
|
Vidal L, Brennan MÁ, Krissian S, De Lima J, Hoornaert A, Rosset P, Fellah BH, Layrolle P. In situ production of pre-vascularized synthetic bone grafts for regenerating critical-sized defects in rabbits. Acta Biomater 2020; 114:384-394. [PMID: 32688088 DOI: 10.1016/j.actbio.2020.07.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/16/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022]
Abstract
Reconstructing large bone defects caused by severe trauma or resection of tumors remains a challenge for surgeons. A fibula free flap and its vascularized bed can be transplanted to the reconstruction site to achieve healing. However, this technique adds morbidity, and requires microsurgery and sculpting of the bone tissue to adapt the graft to both the vasculature and the anatomy of the defect. The aim of the current study was to evaluate an alternative approach consisting of the in situ production of a pre-vascularized synthetic bone graft and its subsequent transplantation to a critical-sized bone defect. 3D printed chambers containing biphasic calcium phosphate (BCP) granules, perfused by a local vascular pedicle, with or without the addition of stromal vascular fraction (SVF), were subcutaneously implanted into New Zealand White female rabbits. SVF was prepared extemporaneously from autologous adipose tissue, the vascular pedicle was isolated from the inguinal site, while BCP granules alone served as a control group. After 8 weeks, the constructs containing a vascular pedicle exhibited abundant neovascularization with blood vessels sprouting from the pedicle, leading to significantly increased vascularization compared to BCP controls. Pre-vascularized synthetic bone grafts were then transplanted into 15 mm critical-sized segmental ulnar defects for a further 8 weeks. Micro-CT and decalcified histology revealed that pre-vascularization of synthetic bone grafts led to enhanced bone regeneration. This pre-clinical study demonstrates the feasibility and efficacy of the in situ production of pre-vascularized synthetic bone grafts for regenerating large bone defects, thereby addressing an important clinical need. STATEMENT OF SIGNIFICANCE: The current gold standard in large bone defect regeneration is vascularized fibula grafting. An alternative approach consisting of in situ production of a pre-vascularized synthetic bone graft and its subsequent transplantation to a bone defect is presented here. 3D printed chambers were filled with biphasic calcium phosphate granules, supplemented with autologous stromal vascular fraction and an axial vascular pedicle and subcutaneously implanted in inguinal sites. These pre-vascularized synthetic grafts were then transplanted into critical-sized segmental ulnar defects. Micro-CT and decalcified histology revealed that the pre-vascularized synthetic bone grafts led to higher bone regeneration than non-vascularized constructs. An alternative to vascularized fibula grafting is provided and may address an important clinical need for large bone defect reconstruction.
Collapse
|
11
|
Chen Y, Shen J, Ma C, Cao M, Yan J, Liang J, Ke K, Cao M, Xiaosu G. Skin-derived precursor Schwann cells protect SH-SY5Y cells against 6-OHDA-induced neurotoxicity by PI3K/AKT/Bcl-2 pathway. Brain Res Bull 2020; 161:84-93. [PMID: 32360763 DOI: 10.1016/j.brainresbull.2020.03.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 03/22/2020] [Accepted: 03/31/2020] [Indexed: 02/08/2023]
Abstract
Skin-derived precursors (SKPs) are self-renewing and pluripotent adult stem cell sources that have been successfully obtained and cultured from adult tissues of rodents and humans. Skin-derived precursor Schwann cells (SKP-SCs), derived from SKPs when cultured in a neuro stromal medium supplemented with some appropriate neurotrophic factors, have been reported to play a neuroprotective effect in the peripheral nervous system. This proves our previous studies that SKP-SCs' function to bridge sciatic nerve gap in rats. However, the function of SKP-SCs in Parkinson disease (PD) remains unknown. This study was aimed to investigate the possible neuroprotective effects of SKP-SCs in 6-OHDA-induced Parkinson's disease (PD) model. Our results showed that the treatment with SKP-SCs prevented SH-SY5Y cells from 6-OHDA-induced apoptosis, accompanied by modulation of apoptosis-related proteins (Bcl-2 and Bax) and the decreased expression of active caspase-3. Furthermore, we confirmed that SKP-SCs might exert protective effects and increase the mitochondrial membrane potential (MMP) through PI3K/AKT/Bcl-2 pathway. Taken together, our results demonstrated that SKP-SCs protect against 6-OHDA-induced cytotoxicity through PI3K/AKT/Bcl-2 pathway in PD model in vitro, which provides a new theoretical basis for the treatment of PD.
Collapse
Affiliation(s)
- Ying Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Chengxiao Ma
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Maosheng Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jianan Yan
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jingjing Liang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China.
| | - Gu Xiaosu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China.
| |
Collapse
|
12
|
Regeneration of segmental defects in metatarsus of sheep with vascularized and customized 3D-printed calcium phosphate scaffolds. Sci Rep 2020; 10:7068. [PMID: 32341459 PMCID: PMC7184564 DOI: 10.1038/s41598-020-63742-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/31/2020] [Indexed: 11/12/2022] Open
Abstract
Although autografts are considered to be the gold standard treatment for reconstruction of large bone defects resulting from trauma or diseases, donor site morbidity and limited availability restrict their use. Successful bone repair also depends on sufficient vascularization and to address this challenge, novel strategies focus on the development of vascularized biomaterial scaffolds. This pilot study aimed to investigate the feasibility of regenerating large bone defects in sheep using 3D-printed customized calcium phosphate scaffolds with or without surgical vascularization. Pre-operative computed tomography scans were performed to visualize the metatarsus and vasculature and to fabricate customized scaffolds and surgical guides by 3D printing. Critical-sized segmental defects created in the mid-diaphyseal region of the metatarsus were either left empty or treated with the 3D scaffold alone or in combination with an axial vascular pedicle. Bone regeneration was evaluated 1, 2 and 3 months post-implantation. After 3 months, the untreated defect remained non-bridged while the 3D scaffold guided bone regeneration. The presence of the vascular pedicle further enhanced bone formation. Histology confirmed bone growth inside the porous 3D scaffolds with or without vascular pedicle inclusion. Taken together, this pilot study demonstrated the feasibility of precised pre-surgical planning and reconstruction of large bone defects with 3D-printed personalized scaffolds.
Collapse
|
13
|
Marolt Presen D, Traweger A, Gimona M, Redl H. Mesenchymal Stromal Cell-Based Bone Regeneration Therapies: From Cell Transplantation and Tissue Engineering to Therapeutic Secretomes and Extracellular Vesicles. Front Bioeng Biotechnol 2019; 7:352. [PMID: 31828066 PMCID: PMC6890555 DOI: 10.3389/fbioe.2019.00352] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Effective regeneration of bone defects often presents significant challenges, particularly in patients with decreased tissue regeneration capacity due to extensive trauma, disease, and/or advanced age. A number of studies have focused on enhancing bone regeneration by applying mesenchymal stromal cells (MSCs) or MSC-based bone tissue engineering strategies. However, translation of these approaches from basic research findings to clinical use has been hampered by the limited understanding of MSC therapeutic actions and complexities, as well as costs related to the manufacturing, regulatory approval, and clinical use of living cells and engineered tissues. More recently, a shift from the view of MSCs directly contributing to tissue regeneration toward appreciating MSCs as "cell factories" that secrete a variety of bioactive molecules and extracellular vesicles with trophic and immunomodulatory activities has steered research into new MSC-based, "cell-free" therapeutic modalities. The current review recapitulates recent developments, challenges, and future perspectives of these various MSC-based bone tissue engineering and regeneration strategies.
Collapse
Affiliation(s)
- Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andreas Traweger
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Spinal Cord Injury & Tissue Regeneration Center Salzburg, Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
14
|
Sabater González M, Calvo Carrasco D. Advances in Exotic Animal Osteosynthesis. Vet Clin North Am Exot Anim Pract 2019; 22:441-450. [PMID: 31395324 DOI: 10.1016/j.cvex.2019.06.006] [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: 10/26/2022]
Abstract
Exotic animal orthopedics has not incorporated the most recent progress made in small animal surgery or human medicine. Although minimally invasive osteosynthesis has been incorporated as a routinely used alternative in small animals, its use in exotic animals is still in its infancy. This article compliments the reviews of orthopedics in small mammals, birds, and reptiles in the previous issue. It reviews relevant recent studies performed in laboratory animals about new orthopedic materials and techniques showing potential to become incorporated into the routine orthopedic treatment of exotic animals in the coming years.
Collapse
|
15
|
Wang Z, Mithieux SM, Weiss AS. Fabrication Techniques for Vascular and Vascularized Tissue Engineering. Adv Healthc Mater 2019; 8:e1900742. [PMID: 31402593 DOI: 10.1002/adhm.201900742] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/12/2019] [Indexed: 12/19/2022]
Abstract
Impaired or damaged blood vessels can occur at all levels in the hierarchy of vascular systems from large vasculatures such as arteries and veins to meso- and microvasculatures such as arterioles, venules, and capillary networks. Vascular tissue engineering has become a promising approach for fabricating small-diameter vascular grafts for occlusive arteries. Vascularized tissue engineering aims to fabricate meso- and microvasculatures for the prevascularization of engineered tissues and organs. The ideal small-diameter vascular graft is biocompatible, bridgeable, and mechanically robust to maintain patency while promoting tissue remodeling. The desirable fabricated meso- and microvasculatures should rapidly integrate with the host blood vessels and allow nutrient and waste exchange throughout the construct after implantation. A number of techniques used, including engineering-based and cell-based approaches, to fabricate these synthetic vasculatures are herein explored, as well as the techniques developed to fabricate hierarchical structures that comprise multiple levels of vasculature.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Suzanne M. Mithieux
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Anthony S. Weiss
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
- Bosch Institute University of Sydney NSW 2006 Australia
- Sydney Nano Institute University of Sydney NSW 2006 Australia
| |
Collapse
|
16
|
Mohiuddin OA, Campbell B, Poche JN, Ma M, Rogers E, Gaupp D, Harrison MAA, Bunnell BA, Hayes DJ, Gimble JM. Decellularized Adipose Tissue Hydrogel Promotes Bone Regeneration in Critical-Sized Mouse Femoral Defect Model. Front Bioeng Biotechnol 2019; 7:211. [PMID: 31552237 PMCID: PMC6743019 DOI: 10.3389/fbioe.2019.00211] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 08/19/2019] [Indexed: 12/18/2022] Open
Abstract
Critical-sized bone defects fail to heal and often cause non-union. Standard treatments employ autologous bone grafting, which can cause donor tissue loss/pain. Although several scaffold types can enhance bone regeneration, multiple factors limit their level of success. To address this issue, this study evaluated a novel decellularized human adipose tissue (DAT) hydrogel as an alternative. In this study, DAT hydrogel alone, or in combination with adipose-derived stromal/stem cells (ASC), osteo-induced ASCs (OIASC), and hydroxyapatite were tested for their ability to mediate repair of a critical-sized (3 mm) femoral defect created in C57BL/6 mice. Micro-computed tomography results showed that all DAT hydrogel treated groups significantly enhanced bone regeneration, with OIASC + hydroxyapatite treated group displaying the most robust bone regeneration. Histological analyses revealed that all treatments resulted in significantly higher tissue areas with the relative mineralized tissue area significantly increased at 12 weeks; however, cartilaginous content was lowest among treatment groups with OIASC. Immunohistochemical analyses showed that DAT hydrogel enhanced collagen I and osteopontin expression, while the addition of OIASCs to the hydrogel reduced collagen II levels. Thus, DAT hydrogel promotes bone regeneration in a critical-sized femoral defect model that is further enhanced in the presence of OIASCs and hydroxyapatite.
Collapse
Affiliation(s)
- Omair A Mohiuddin
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Brett Campbell
- School of Medicine, Tulane University, New Orleans, LA, United States
| | - J Nick Poche
- School of Medicine, Louisiana State University, New Orleans, LA, United States
| | - Michelle Ma
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, United States.,Obatala Sciences, New Orleans, LA, United States
| | - Emma Rogers
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, United States
| | - Dina Gaupp
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Mark A A Harrison
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Daniel J Hayes
- Department of Biomedical Engineering, Pennsylvania State University, State College, PA, United States
| | - Jeffrey M Gimble
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, United States.,LaCell LLC, New Orleans, LA, United States
| |
Collapse
|
17
|
Ahangar P, Aziz M, Rosenzweig DH, Weber MH. Advances in personalized treatment of metastatic spine disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:223. [PMID: 31297388 DOI: 10.21037/atm.2019.04.41] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The spine is one of the most common sites of bony metastases, and its involvement leads to significant patient morbidity. Surgical management in these patients is aimed at improving quality of life and functional status throughout the course of the disease. Resection of metastases often leads to critical size bone defects, presenting a challenge to achieving adequate bone regeneration to fill the void. Current treatment options for repairing these defects are bone grafting and commercial bone cements; however, each has associated limitations. Additionally, tumor recurrence and tumor-induced bone loss make bone regeneration particularly difficult. Systemic therapeutic delivery, such as bisphosphonates, have become standard of care to combat bone loss despite unfavorable systemic side-effects and lack of local efficacy. Developments from tissue engineering have introduced novel materials with osteoinductive and osteoconductive properties which also act as structural support scaffolds for bone regeneration. These new materials can also act as a therapeutic reservoir to sustainably release drugs locally as an alternative to systemic therapy. In this review, we outline recent advancements in tissue engineering and the role of translational research in developing implants that can fully repair bone defects while also delivering local therapeutics to curb tumor recurrence and improve patient quality of life.
Collapse
Affiliation(s)
- Pouyan Ahangar
- Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada.,The Research Institute of the McGill University Health Centre, Injury, Repair and Recovery Program, Montreal, QC, Canada.,Montreal General Hospital C10.148.6, Montreal, QC, Canada
| | - Mina Aziz
- Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada.,The Research Institute of the McGill University Health Centre, Injury, Repair and Recovery Program, Montreal, QC, Canada.,Montreal General Hospital C10.148.6, Montreal, QC, Canada.,Clinical Investigator Program, McGill University, Montreal, QC, Canada
| | - Derek H Rosenzweig
- Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada.,The Research Institute of the McGill University Health Centre, Injury, Repair and Recovery Program, Montreal, QC, Canada.,Montreal General Hospital C10.148.6, Montreal, QC, Canada
| | - Michael H Weber
- Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada.,The Research Institute of the McGill University Health Centre, Injury, Repair and Recovery Program, Montreal, QC, Canada.,Montreal General Hospital C10.148.6, Montreal, QC, Canada
| |
Collapse
|
18
|
Current Biomedical Applications of 3D Printing and Additive Manufacturing. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9081713] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Additive manufacturing (AM) has emerged over the past four decades as a cost-effective, on-demand modality for fabrication of geometrically complex objects. The ability to design and print virtually any object shape using a diverse array of materials, such as metals, polymers, ceramics and bioinks, has allowed for the adoption of this technology for biomedical applications in both research and clinical settings. Current advancements in tissue engineering and regeneration, therapeutic delivery, medical device fabrication and operative management planning ensure that AM will continue to play an increasingly important role in the future of healthcare. In this review, we outline current biomedical applications of common AM techniques and materials.
Collapse
|
19
|
Liu M, Lv Y. Reconstructing Bone with Natural Bone Graft: A Review of In Vivo Studies in Bone Defect Animal Model. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E999. [PMID: 30513940 PMCID: PMC6315600 DOI: 10.3390/nano8120999] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 11/25/2018] [Accepted: 11/29/2018] [Indexed: 12/28/2022]
Abstract
Bone defects caused by fracture, disease or congenital defect remains a medically important problem to be solved. Bone tissue engineering (BTE) is a promising approach by providing scaffolds to guide and support the treatment of bone defects. However, the autologous bone graft has many defects such as limited sources and long surgical procedures. Therefore, xenograft bone graft is considered as one of the best substitutions and has been effectively used in clinical practice. Due to better preserved natural bone structure, suitable mechanical properties, low immunogenicity, good osteoinductivity and osteoconductivity in natural bone graft, decellularized and demineralized bone matrix (DBM) scaffolds were selected and discussed in the present review. In vivo animal models provide a complex physiological environment for understanding and evaluating material properties and provide important reference data for clinical trials. The purpose of this review is to outline the in vivo bone regeneration and remodeling capabilities of decellularized and DBM scaffolds in bone defect models to better evaluate the potential of these two types of scaffolds in BTE. Taking into account the limitations of the state-of-the-art technology, the results of the animal bone defect model also provide important information for future design of natural bone composite scaffolds.
Collapse
Affiliation(s)
- Mengying Liu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, China.
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
20
|
Zhu C, Huang J, Xue C, Wang Y, Wang S, Bao S, Chen R, Li Y, Gu Y. Skin derived precursor Schwann cell-generated acellular matrix modified chitosan/silk scaffolds for bridging rat sciatic nerve gap. Neurosci Res 2018; 135:21-31. [DOI: 10.1016/j.neures.2017.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/24/2017] [Accepted: 12/25/2017] [Indexed: 12/12/2022]
|
21
|
Abstract
Bioreactors have become indispensable tools in the cell-based therapy industry. Various forms of bioreactors are used to maintain well-controlled microenvironments to regulate cell growth, differentiation, and tissue development. They are essential for providing standardized, reproducible cell-based products for regenerative medicine applications or to establish physiologically relevant
in vitro models for testing of pharmacologic agents. In this review, we discuss three main classes of bioreactors: cell expansion bioreactors, tissue engineering bioreactors, and lab-on-a-chip systems. We briefly examine the factors driving concerted research endeavors in each of these areas and describe the major advancements that have been reported in the last three years. Emerging issues that impact the commercialization and clinical use of bioreactors include (i) the need to scale up to greater cell quantities and larger graft sizes, (ii) simplification of
in vivo systems to function without exogenous stem cells or growth factors or both, and (iii) increased control in the manufacture and monitoring of miniaturized systems to better capture complex tissue and organ physiology.
Collapse
Affiliation(s)
- Makeda Stephenson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Warren Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Ismail T, Osinga R, Todorov A, Haumer A, Tchang LA, Epple C, Allafi N, Menzi N, Largo RD, Kaempfen A, Martin I, Schaefer DJ, Scherberich A. Engineered, axially-vascularized osteogenic grafts from human adipose-derived cells to treat avascular necrosis of bone in a rat model. Acta Biomater 2017; 63:236-245. [PMID: 28893630 DOI: 10.1016/j.actbio.2017.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/04/2017] [Accepted: 09/05/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Avascular necrosis of bone (AVN) leads to sclerosis and collapse of bone and joints. The standard of care, vascularized bone grafts, is limited by donor site morbidity and restricted availability. The aim of this study was to generate and test engineered, axially vascularized SVF cells-based bone substitutes in a rat model of AVN. METHODS SVF cells were isolated from lipoaspirates and cultured onto porous hydroxyapatite scaffolds within a perfusion-based bioreactor system for 5days. The resulting constructs were inserted into devitalized bone cylinders mimicking AVN-affected bone. A ligated vascular bundle was inserted upon subcutaneous implantation of constructs in nude rats. After 1 and 8weeks in vivo, bone formation and vascularization were analyzed. RESULTS Newly-formed bone was found in 80% of SVF-seeded scaffolds after 8weeks but not in unseeded controls. Human ALU+cells in the bone structures evidenced a direct contribution of SVF cells to bone formation. A higher density of regenerative, M2 macrophages was observed in SVF-seeded constructs. In both experimental groups, devitalized bone was revitalized by vascularized tissue after 8 weeks. CONCLUSION SVF cells-based osteogenic constructs revitalized fully necrotic bone in a challenging AVN rat model of clinically-relevant size. SVF cells contributed to accelerated initial vascularization, to bone formation and to recruitment of pro-regenerative endogenous cells. STATEMENT OF SIGNIFICANCE Avascular necrosis (AVN) of bone often requires surgical treatment with autologous bone grafts, which is surgically demanding and restricted by significant donor site morbidity and limited availability. This paper describes a de novo engineered axially-vascularized bone graft substitute and tests the potential to revitalize dead bone and provide efficient new bone formation in a rat model. The engineering of an osteogenic/vasculogenic construct of clinically-relevant size with stromal vascular fraction of human adipose, combined to an arteriovenous bundle is described. This construct revitalized and generated new bone tissue. This successful approach proposes a novel paradigm in the treatment of AVN, in which an engineered, vascularized osteogenic graft would be used as a germ to revitalize large volumes of necrotic bone.
Collapse
|
23
|
Jalili-Firoozinezhad S, Martin I, Scherberich A. Bimodal morphological analyses of native and engineered tissues. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:543-550. [DOI: 10.1016/j.msec.2017.03.140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 12/23/2022]
|
24
|
Contrast-Enhanced Microtomographic Characterisation of Vessels in Native Bone and Engineered Vascularised Grafts Using Ink-Gelatin Perfusion and Phosphotungstic Acid. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:4035160. [PMID: 29097920 PMCID: PMC5612680 DOI: 10.1155/2017/4035160] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/18/2017] [Accepted: 04/02/2017] [Indexed: 11/17/2022]
Abstract
Objectives Bone ischemia and necrosis are challenging to treat, requiring investigation of native and engineered bone revascularisation processes through advanced imaging techniques. This study demonstrates an experimental two-step method for precise bone and vessel analysis in native bones or vascularised bone grafts using X-ray microtomography (μCT), without interfering with further histological processing. Methods Distally ligated epigastric arteries or veins of 6 nude rats were inserted in central channels of porous hydroxyapatite cylinders and these pedicled grafts were implanted subcutaneously. One week later, the rats were perfused with ink-gelatin and euthanised and the femurs, tibias, and grafts were explanted. Samples were scanned using μCT, decalcified, incubated with phosphotungstic acid (PTA) for contrast enhancement, rescanned, and processed histologically. Results Contrast-enhanced μCT displayed the course and branching of native bone vessels. Histologically, both central (-17%) and epiphyseal vessels (-58%) appeared smaller than in μCT scans. Hydroxyapatite cylinders were thoroughly vascularised but did not display bone formation. Grafts with a central artery had more (+58%) and smaller (-52%) vessel branches compared to grafts with a vein. Conclusions We present a relatively inexpensive and easy-to-perform two-step method to analyse bone and vessels by μCT, suitable to assess a variety of bone-regenerative strategies.
Collapse
|
25
|
Nakamura N, Kimura T, Kishida A. Overview of the Development, Applications, and Future Perspectives of Decellularized Tissues and Organs. ACS Biomater Sci Eng 2016; 3:1236-1244. [DOI: 10.1021/acsbiomaterials.6b00506] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Naoko Nakamura
- Institute of Biomaterials
and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062 Japan
| | - Tsuyoshi Kimura
- Institute of Biomaterials
and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062 Japan
| | - Akio Kishida
- Institute of Biomaterials
and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062 Japan
| |
Collapse
|
26
|
Kagami H. Potential application of tissue engineering for the reconstruction of facial bones. Oral Dis 2016; 23:689-691. [DOI: 10.1111/odi.12581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- H Kagami
- Department of Oral and Maxillofacial Surgery; School of Dentistry; Matsumoto Dental University; Shiojiri
- Department of Advanced Medical Science; IMSUT Hospital; The Institute of Medical Science; The University of Tokyo; Tokyo Japan
| |
Collapse
|
27
|
Laschke MW, Menger MD. Prevascularization in tissue engineering: Current concepts and future directions. Biotechnol Adv 2015; 34:112-21. [PMID: 26674312 DOI: 10.1016/j.biotechadv.2015.12.004] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/16/2015] [Accepted: 12/04/2015] [Indexed: 12/24/2022]
Abstract
The survival of engineered tissue constructs during the initial phase after their implantation depends on the rapid development of an adequate vascularization. This, in turn, is a major prerequisite for the constructs' long-term function. 'Prevascularization' has emerged as a promising concept in tissue engineering, aiming at the generation of a preformed microvasculature in tissue constructs prior to their implantation. This should shorten the time period during which the constructs are avascular and suffer hypoxic conditions. Herein, we provide an overview of current strategies for the generation of preformed microvascular networks within tissue constructs. In vitro approaches use cell seeding, spheroid formation or cell sheet technologies. In situ approaches use the body as a natural bioreactor to induce vascularization by angiogenic ingrowth or flap and arteriovenous (AV)-loop techniques. In future, these strategies may be supplemented by the transplantation of adipose tissue-derived microvascular fragments or the in vitro generation of highly organized microvascular networks by means of sophisticated microscale technologies and microfluidic systems. The further advancement of these prevascularization concepts and their adaptation to individual therapeutic interventions will markedly contribute to a broad implementation of tissue engineering applications into clinical practice.
Collapse
Affiliation(s)
- Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, D-66421 Homburg/Saar, Germany.
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, D-66421 Homburg/Saar, Germany
| |
Collapse
|