1
|
Ghezzi B, Matera B, Meglioli M, Rossi F, Duraccio D, Faga MG, Zappettini A, Macaluso GM, Lumetti S. Composite PCL Scaffold With 70% β-TCP as Suitable Structure for Bone Replacement. Int Dent J 2024; 74:1220-1232. [PMID: 38614878 DOI: 10.1016/j.identj.2024.02.013] [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: 12/10/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 04/15/2024] Open
Abstract
OBJECTIVES The purpose of this work was to optimise printable polycaprolactone (PCL)/β-tricalcium phosphate (β-TCP) biomaterials with high percentages of β-TCP endowed with balanced mechanical characteristics to resemble human cancellous bone, presumably improving osteogenesis. METHODS PCL/β-TCP scaffolds were obtained from customised filaments for fused deposition modelling (FDM) 3D printing with increasing amounts of β-TCP. Samples mechanical features, surface topography and wettability were evaluated as well as cytocompatibility assays, cell adhesion and differentiation. RESULTS The parameters of the newly fabricated materila were optimal for PCL/β-TCP scaffold fabrication. Composite surfaces showed higher hydrophilicity compared with the controls, and their surface roughness sharply was higher, possibly due to the presence of β-TCP. The Young's modulus of the composites was significantly higher than that of pristine PCL, indicating that the intrinsic strength of β-TCP is beneficial for enhancing the elastic modulus of the composite biomaterials. All novel composite biomaterials supported greater cellular growth and stronger osteoblastic differentiation compared with the PCL control. CONCLUSIONS This project highlights the possibility to fabricat, through an FDM solvent-free approach, PCL/β-TCP scaffolds of up to 70 % concentrations of β-TCP. overcoming the current lmit of 60 % stated in the literature. The combination of 3D printing and customised biomaterials allowed production of highly personalised scaffolds with optimal mechanical and biological features resembling the natural structure and the composition of bone. This underlines the promise of such structures for innovative approaches for bone and periodontal regeneration.
Collapse
Affiliation(s)
- Benedetta Ghezzi
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy; Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Biagio Matera
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
| | - Matteo Meglioli
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy.
| | - Francesca Rossi
- Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Donatella Duraccio
- Istituto di Scienze e Tecnologie per l'Energia e la Mobilità Sostenibili, Consiglio Nazionale delle Ricerche, Torino, Italy
| | - Maria Giulia Faga
- Istituto di Scienze e Tecnologie per l'Energia e la Mobilità Sostenibili, Consiglio Nazionale delle Ricerche, Torino, Italy
| | - Andrea Zappettini
- Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Guido Maria Macaluso
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy; Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Simone Lumetti
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy; Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| |
Collapse
|
2
|
Ximenes-Carballo C, Rey-Viñolas S, Blanco-Fernandez B, Pérez-Amodio S, Engel E, Castano O. Combining three-dimensionality and CaP glass-PLA composites: Towards an efficient vascularization in bone tissue healing. BIOMATERIALS ADVANCES 2024; 164:213985. [PMID: 39146606 DOI: 10.1016/j.bioadv.2024.213985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Bone regeneration often fails due to implants/grafts lacking vascular supply, causing necrotic tissue and poor integration. Microsurgical techniques are used to overcome this issue, allowing the graft to anastomose. These techniques have limitations, including severe patient morbidity and current research focuses on stimulating angiogenesis in situ using growth factors, presenting limitations, such as a lack of control and increased costs. Non-biological stimuli are necessary to promote angiogenesis for successful bone constructs. Recent studies have reported that bioactive glass dissolution products, such as calcium-releasing nanoparticles, stimulate hMSCs to promote angiogenesis and new vasculature. Moreover, the effect of 3D microporosity has also been reported to be important for vascularisation in vivo. Therefore, we used room-temperature extrusion 3D printing with polylactic acid (PLA) and calcium phosphate (CaP) based glass scaffolds, focusing on geometry and solvent displacement for scaffold recovery. Combining both methods enabled reproducible control of 3D structure, porosity, and surface topography. Scaffolds maintained calcium ion release at physiological levels and supported human mesenchymal stem cell proliferation. Scaffolds stimulated the secretion of vascular endothelial growth factor (VEGF) after 3 days of culture. Subcutaneous implantation in vivo indicated good scaffold integration and blood vessel infiltration as early as one week after. PLA-CaP scaffolds showed increased vessel maturation 4 weeks after implantation without vascular regression. Results show PLA/CaP-based glass scaffolds, made via controlled 3D printing, support angiogenesis and vessel maturation, promising improved vascularization for bone regeneration.
Collapse
Affiliation(s)
- Celia Ximenes-Carballo
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Sergi Rey-Viñolas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Barbara Blanco-Fernandez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Soledad Pérez-Amodio
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; IMEM-BRT group, Materials Science and Engineering, Polytechnical University of Catalonia (UPC), Barcelona, Spain
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; IMEM-BRT group, Materials Science and Engineering, Polytechnical University of Catalonia (UPC), Barcelona, Spain.
| | - Oscar Castano
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain; Nanobioengineering and Biomaterials, Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.
| |
Collapse
|
3
|
Earley J, Roebuck D. Amputation and limb-sparing surgery in childhood sarcoma: Post-operative imaging appearances and complications. J Med Imaging Radiat Oncol 2024. [PMID: 39428120 DOI: 10.1111/1754-9485.13790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/22/2024] [Indexed: 10/22/2024]
Abstract
Limb-sparing surgery and amputation are common surgical techniques used to achieve local tumour control in childhood primary bone and soft tissue malignancy of the limbs. The interpretation of post-operative limb imaging in these frequently complex cases is assisted by knowledge of the surgical techniques employed. This review discusses the rationale underpinning the most common surgical techniques used for these patients as well as their expected post-operative imaging appearance and complications. Amputation, long bone resection, endoprosthetic reconstruction, allograft reconstruction, the use of fibular autografts, allograft-prosthetic composite reconstruction and arthrodesis are discussed.
Collapse
Affiliation(s)
- Joel Earley
- Western Australian Radiology Training Program, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Derek Roebuck
- Perth Children's Hospital, Nedlands, Australia, Australia
- University of Western Australia, Crawley, Australia, Australia
| |
Collapse
|
4
|
Manon J, Englebert A, Evrard R, Schubert T, Cornu O. FixThePig: a custom 3D-printed femoral intramedullary nailing for preclinical research applications. Front Bioeng Biotechnol 2024; 12:1478676. [PMID: 39493302 PMCID: PMC11528544 DOI: 10.3389/fbioe.2024.1478676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
Background Critical-size bone defects (CSBDs) pose significant challenges in clinical orthopaedics and traumatology. Developing reliable preclinical models that accurately simulate human conditions is crucial for translational research. This study addresses the need for a reliable preclinical model by evaluating the design and efficacy of a custom-made 3D-printed intramedullary nail (IMN) specifically for CSBDs in minipigs. The study aims to answer the following questions: Can a custom-made 3D-printed IMN be designed for femoral osteosynthesis in minipigs? Does the use of the custom-made IMN result in consistent and reproducible surgical procedure, particularly in the creation and fixation of CSBDs? Can the custom-made IMN effectively treat and promote bone consolidation of CSBDs? Hypothesis The custom-made 3D-printed IMN can be designed to effectively create, fix and treat CSBDs in minipigs, resulting in consistent surgical outcomes. Materials and Methods The IMN was designed based on CT scans of minipig femurs, considering factors such as femoral curvature, length, and medullary canal diameters. It was 3D-printed in titanium and evaluated through both in vitro and in vivo testing. Female Aachen minipigs underwent bilateral femoral surgeries to create and fix CSBDs using the custom-made IMN. Post-operative follow-up included X-rays and CT scans every 2 weeks, with manual examination of explanted femurs to assess consolidation and mechanical stability after 3 months. Results The custom-made IMN effectively fitted the minipig femoral anatomy and facilitated reproducible surgical outcomes. Symmetric double osteotomies were successfully performed, and allografts showed minimal morphological discrepancies. However, proximal fixation faced challenges, leading to non-union in several cases, while most distal osteotomy sites achieved stable consolidation. Discussion The custom-made 3D-printed IMN demonstrated potential in modelling and treating CSBDs in minipigs. While the design effectively supported distal bone healing, issues with proximal fixation highlight the need for further refinements. Potential improvements include better screw placement, additional mechanical support, and adaptations such as a reduction clamp or a cephalic screw to enhance stability and distribute forces more effectively.
Collapse
Affiliation(s)
- Julie Manon
- Neuro Musculo Skeletal Lab (NMSK), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Bruxelles, Belgium
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
- Unité de Thérapie Tissulaire et Cellulaire de l’Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
| | - Alexandre Englebert
- Neuro Musculo Skeletal Lab (NMSK), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Bruxelles, Belgium
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
- Institute for Information and Communication Technologies, Electronics and Applied Mathematics (ICTEAM), Electrical Engineering Department (ELEN), UCLouvain, Louvain-la-Neuve, Belgium
| | - Robin Evrard
- Neuro Musculo Skeletal Lab (NMSK), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Bruxelles, Belgium
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
- Unité de Thérapie Tissulaire et Cellulaire de l’Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
| | - Thomas Schubert
- Neuro Musculo Skeletal Lab (NMSK), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Bruxelles, Belgium
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
- Unité de Thérapie Tissulaire et Cellulaire de l’Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
| | - Olivier Cornu
- Neuro Musculo Skeletal Lab (NMSK), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Bruxelles, Belgium
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
- Unité de Thérapie Tissulaire et Cellulaire de l’Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
| |
Collapse
|
5
|
Cheers GM, Weimer LP, Neuerburg C, Arnholdt J, Gilbert F, Thorwächter C, Holzapfel BM, Mayer-Wagner S, Laubach M. Advances in implants and bone graft types for lumbar spinal fusion surgery. Biomater Sci 2024; 12:4875-4902. [PMID: 39190323 DOI: 10.1039/d4bm00848k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The increasing prevalence of spinal disorders worldwide necessitates advanced treatments, particularly interbody fusion for severe cases that are unresponsive to non-surgical interventions. This procedure, especially 360° lumbar interbody fusion, employs an interbody cage, pedicle screw-and-rod instrumentation, and autologous bone graft (ABG) to enhance spinal stability and promote fusion. Despite significant advancements, a persistent 10% incidence of non-union continues to result in compromised patient outcomes and escalated healthcare costs. Innovations in lumbar stabilisation seek to mimic the properties of natural bone, with evolving implant materials like titanium (Ti) and polyetheretherketone (PEEK) and their composites offering new prospects. Additionally, biomimetic cages featuring precisely engineered porosities and interconnectivity have gained traction, as they enhance osteogenic differentiation, support osteogenesis, and alleviate stress-shielding. However, the limitations of ABG, such as harvesting morbidities and limited fusion capacity, have spurred the exploration of sophisticated solutions involving advanced bone graft substitutes. Currently, demineralised bone matrix and ceramics are in clinical use, forming the basis for future investigations into novel bone graft substitutes. Bioglass, a promising newcomer, is under investigation despite its observed rapid absorption and the potential for foreign body reactions in preclinical studies. Its clinical applicability remains under scrutiny, with ongoing research addressing challenges related to burst release and appropriate dosing. Conversely, the well-documented favourable osteogenic potential of growth factors remains encouraging, with current efforts focused on modulating their release dynamics to minimise complications. In this evidence-based narrative review, we provide a comprehensive overview of the evolving landscape of non-degradable spinal implants and bone graft substitutes, emphasising their applications in lumbar spinal fusion surgery. We highlight the necessity for continued research to improve clinical outcomes and enhance patient well-being.
Collapse
Affiliation(s)
- Giles Michael Cheers
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany.
| | - Lucas Philipp Weimer
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany.
| | - Carl Neuerburg
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany.
| | - Jörg Arnholdt
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany.
| | - Fabian Gilbert
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany.
| | - Christoph Thorwächter
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany.
| | - Boris Michael Holzapfel
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany.
| | - Susanne Mayer-Wagner
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany.
| | - Markus Laubach
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany.
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
| |
Collapse
|
6
|
Zhang R, Yao X, Wang X, Zheng X, Mu H, Ruan H, Kang Q. Calcaneal Lengthening after Tarsal Bone Fusion for Massive Calcaneus Defect Reconstruction. Orthop Surg 2024. [PMID: 39235036 DOI: 10.1111/os.14230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 09/06/2024] Open
Abstract
OBJECTIVES Calcaneus defect remains challenging with limited strategies for reconstruction. Current methods, including graft transplantation, substitution, and distraction osteogenesis, showed limited advantages with certain shortcomings. Current calcaneus lengthening for partial calcaneus loss reconstruction requires bone loss of less than 35%. We introduced our combination of tarsal bone fusion and gradual lengthening method in treating massive calcaneus loss. METHODS From January 2015 to December 2021, tarsal bone fusion and calcaneus gradual lengthening were performed in six patients with unilateral massive traumatic loss of the calcaneal tuberosity. A retrospective study was held to evaluate the outcomes of this novel technique. Clinical outcomes were assessed based on the American Orthopedic Foot and Ankle Score (AOFAS). Radiological data were assessed, which included tibio-calcaneal angle (TCA), calcaneal interface angle (CIA), metatarsal declination angle (MDA), angle of longitudinal arch (ALA), and the amount of calcaneus axial lengthening (CAL). RESULTS The mean calcaneal axial lengthening was 43.8 ± 3.1 mm (range, 39-49.5 mm), and the mean proportion of the lengthened calcaneus was 47.8% ± 3.7% (range, 42.8-55.3%). The mean external fixation time was 104.8 ± 67.5 days (range, 69 to 242 days), and the mean external fixation index was 2.4 ± 1.6 days/cm. All patients stuck to the postoperative follow-up plan with an average follow-up time (FT) of 35.0 ± 6.7 months (range, 26-40 months). Deformities of the injured limbs were all corrected according to radiography. Based on the AOFAS, three excellent and three good results were achieved. CONCLUSION The Ilizarov technique remains an option for calcaneus reconstruction with a great amount of loss once combined with tarsal bone fusion. The function of the injured foot and ankle can be satisfactorily restored using these techniques in our study. Apart from calcaneus elongation, tarsal bone fusion is somehow necessary to reinforce the proximal segment of the distracted calcaneus for creating a larger distraction callus, correcting concomitant foot deformities, and enhancing hindfoot stability. It is necessary to choose flexibly when tarsal bones should be fused.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangyun Yao
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyu Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu Zheng
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoran Mu
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjiang Ruan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qinglin Kang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Astudillo Potes MD, Mitra I, Hanson K, Camilleri ET, Gaihre B, Shafi M, Hamouda A, Lu L, Elder BD. Biodegradable poly(caprolactone fumarate) 3D printed scaffolds for segmental bone defects within the Masquelet technique. J Orthop Res 2024; 42:1974-1983. [PMID: 38522018 DOI: 10.1002/jor.25839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/21/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024]
Abstract
Segmental bone defects, often clinically treated with nondegradable poly(methylmethacrylate) (PMMA) in multistage surgeries, present a significant clinical challenge. Our study investigated the efficacy of 3D printed biodegradable polycaprolactone fumarate (PCLF)/PCL spacers in a one-stage surgical intervention for these defects, focusing on early bone regeneration influenced by spacer porosities. We compared nonporous PCLF/PCL and PMMA spacers, conventionally molded into cylinders, with porous PCLF/PCL spacers, 3D printed to structurally mimic segmental defects in rat femurs for a 4-week implantation study. Histological analysis, including tissue staining and immunohistochemistry with bone-specific antibodies, was conducted for histomorphometry evaluation. The PCLF/PCL spacers demonstrated compressive properties within 6 ± 0.5 MPa (strength) and 140 ± 15 MPa (modulus). Both porous PCLF/PCL and Nonporous PMMA formed collagen-rich membranes (PCLF/PCL: 92% ± 1.3%, PMMA: 86% ± 1.5%) similar to those induced in the Masquelet technique, indicating PCLF/PCL's potential for one-stage healing. Immunohistochemistry confirmed biomarkers for tissue regeneration, underscoring PCLF/PCL's regenerative capabilities. This research highlights PCLF/PCL scaffolds' ability to induce membrane formation in critical-sized segmental bone defects, supporting their use in one-stage surgery. Both solid and porous PCLF/PCL spacers showed adequate compressive properties, with the porous variants exhibiting BMP-2 expression and woven bone formation, akin to clinical standard PMMA. Notably, the early ossification of the membrane into the pores of porous scaffolds suggests potential for bone interlocking and regeneration, potentially eliminating the need for a second surgery required for PMMA spacers. The biocompatibility and biodegradability of PCLF/PCL make them promising alternatives for treating critical bone defects, especially in vulnerable patient groups.
Collapse
Affiliation(s)
- Maria D Astudillo Potes
- Mayo Clinic Alix School of Medicine, Rochester, Minnesota, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Indranath Mitra
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Kari Hanson
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Emily T Camilleri
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mahnoor Shafi
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Benjamin D Elder
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Regmi A, Niraula BB, Maheshwari V, Nongdamba H, Karn R, Bondarde P, Anand U, Dhingra M, Kandwal P. Establishing a bone bank within a hospital setting in India: early insights from a tertiary care center in Northern India-a review article. Cell Tissue Bank 2024; 25:873-882. [PMID: 39014032 DOI: 10.1007/s10561-024-10146-z] [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: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024]
Abstract
When addressing bone defects resulting from trauma, infection, or tumors, the use of allogenic bone is often necessary. While autografts are considered the standard, they have limitations and can lead to donor site morbidity. Consequently, there has been exploration into the feasibility of utilizing allogenic bone and bone graft replacements. Allogenic bone transplants are acquired from donors following rigorous procurement, sterile processing, and donor screening procedures. To ensure the safe storage and effective utilization of allograft material, a bone banking system is employed. Establishing and managing an orthopedic bone bank, entails navigating complex legal and medical organizational aspects. This paper examines the establishment and operation of bone banks in India, drawing upon our first-hand experience in managing one at a tertiary care center in Northern India.Level of evidence: Level IV.
Collapse
Affiliation(s)
- Anil Regmi
- All India Institute of Medical Sciences, Rishikesh, India
| | | | | | | | - Rahul Karn
- All India Institute of Medical Sciences, Rishikesh, India
| | | | - Utsav Anand
- All India Institute of Medical Sciences, Rishikesh, India
| | - Mohit Dhingra
- All India Institute of Medical Sciences, Rishikesh, India.
| | - Pankaj Kandwal
- All India Institute of Medical Sciences, Rishikesh, India
| |
Collapse
|
9
|
Zhang R, Wang X, Liu S, Qiu S, Ruan H, Kang Q. Modified Calcanization of Tibia for Hindfoot Defect Reconstruction: Method and Preliminary Results. Orthop Surg 2024. [PMID: 39210604 DOI: 10.1111/os.14214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVES To introduce our modified technique for calcanization of the tibia in managing massive bony loss of hindfoot and preliminary outcomes. METHODS From January 2015 to December 2021, modified calcanization of the tibia were performed in 10 patients with unilateral loss of the calcaneus. Clinical outcomes were assessed based on the American Orthopaedic Foot & Ankle Society score and Symptom Checklist-90-Revised questionnaire. Paired two-group t-test was applied to compare the parameters. RESULTS The mean lengthened length of the tibia was 77.3 ± 3.0 mm (range, 74-83 mm). The mean external fixation time was 123.7 ± 52.1 days (range, 117-134 days) and the mean external fixation index was 1.601 ± 0.046 days/mm. All patients stuck to the postoperative follow-up plan with an average follow-up time of 29.7 ± 3.4 months (range, 24-35 months). Deformities of the injured limbs were well corrected. Based on American Orthopaedic Foot & Ankle Society score, eight good and two fair results were achieved. The mental status of all patients was within the normal range, and several indices of the Symptom Checklist-90-Revised questionnaire of each patient were improved after the whole procedure. CONCLUSION We demonstrate that the modified calcanization of the tibia is qualified for total loss of calcaneus with limited complications. Early rehabilitation is attainable since external fixation time is shortened due to a simplified procedure.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyu Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenghe Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Qiu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjiang Ruan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qinglin Kang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Devoy EJ, Jabari E, Kotsanos G, Choe RH, Fisher JP. An Exploration of the Role of Osteoclast Lineage Cells in Bone Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39041616 DOI: 10.1089/ten.teb.2024.0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Bone defects because of age, trauma, and surgery, which are exacerbated by medication side effects and common diseases such as osteoporosis, diabetes, and rheumatoid arthritis, are a problem of epidemic scale. The present clinical standard for treating these defects includes autografts and allografts. Although both treatments can promote robust regenerative outcomes, they fail to strike a desirable balance of availability, side effect profile, consistent regenerative efficacy, and affordability. This difficulty has contributed to the rise of bone tissue engineering (BTE) as a potential avenue through which enhanced bone regeneration could be delivered. BTE is founded upon a paradigm of using biomaterials, bioactive factors, osteoblast lineage cells (ObLCs), and vascularization to cue deficient bone tissue into a state of regeneration. Despite promising preclinical results, BTE has had modest success in being translated into the clinical setting. One barrier has been the simplicity of its paradigm relative to the complexity of biological bone. Therefore, this paradigm must be critically examined and expanded to better account for this complexity. One potential avenue for this is a more detailed consideration of osteoclast lineage cells (OcLCs). Although these cells ostensibly oppose ObLCs and bone regeneration through their resorptive functions, a myriad of investigations have shed light on their potential to influence bone equilibrium in more complex ways through their interactions with both ObLCs and bone matrix. Most BTE research has not systematically evaluated their influence. Yet contrary to expectations associated with the paradigm, a selection of BTE investigations has demonstrated that this influence can enhance bone regeneration in certain contexts. In addition, much work has elucidated the role of many controllable scaffold parameters in both inhibiting and stimulating the activity of OcLCs in parallel to bone regeneration. Therefore, this review aims to detail and explore the implications of OcLCs in BTE and how they can be leveraged to improve upon the existing BTE paradigm.
Collapse
Affiliation(s)
- Eoin J Devoy
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - Erfan Jabari
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - George Kotsanos
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - Robert H Choe
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
11
|
Ashfaq R, Kovács A, Berkó S, Budai-Szűcs M. Developments in Alloplastic Bone Grafts and Barrier Membrane Biomaterials for Periodontal Guided Tissue and Bone Regeneration Therapy. Int J Mol Sci 2024; 25:7746. [PMID: 39062989 PMCID: PMC11277074 DOI: 10.3390/ijms25147746] [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: 06/18/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Periodontitis is a serious form of oral gum inflammation with recession of gingival soft tissue, destruction of the periodontal ligament, and absorption of alveolar bone. Management of periodontal tissue and bone destruction, along with the restoration of functionality and structural integrity, is not possible with conventional clinical therapy alone. Guided bone and tissue regeneration therapy employs an occlusive biodegradable barrier membrane and graft biomaterials to guide the formation of alveolar bone and tissues for periodontal restoration and regeneration. Amongst several grafting approaches, alloplastic grafts/biomaterials, either derived from natural sources, synthesization, or a combination of both, offer a wide variety of resources tailored to multiple needs. Examining several pertinent scientific databases (Web of Science, Scopus, PubMed, MEDLINE, and Cochrane Library) provided the foundation to cover the literature on synthetic graft materials and membranes, devoted to achieving periodontal tissue and bone regeneration. This discussion proceeds by highlighting potential grafting and barrier biomaterials, their characteristics, efficiency, regenerative ability, therapy outcomes, and advancements in periodontal guided regeneration therapy. Marketed and standardized quality products made of grafts and membrane biomaterials have been documented in this work. Conclusively, this paper illustrates the challenges, risk factors, and combination of biomaterials and drug delivery systems with which to reconstruct the hierarchical periodontium.
Collapse
Affiliation(s)
| | | | | | - Mária Budai-Szűcs
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (R.A.); (A.K.); (S.B.)
| |
Collapse
|
12
|
Ji Y, Mao Y, Lin H, Wang Y, Zhao P, Guo Y, Gu L, Fu C, Chen X, Lv Z, Wang N, Li Q, Bei C. Acceleration of bone repairation by BMSCs overexpressing NGF combined with NSA and allograft bone scaffolds. Stem Cell Res Ther 2024; 15:194. [PMID: 38956719 PMCID: PMC11218317 DOI: 10.1186/s13287-024-03807-z] [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: 02/03/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Repairation of bone defects remains a major clinical problem. Constructing bone tissue engineering containing growth factors, stem cells, and material scaffolds to repair bone defects has recently become a hot research topic. Nerve growth factor (NGF) can promote osteogenesis of bone marrow mesenchymal stem cells (BMSCs), but the low survival rate of the BMSCs during transplantation remains an unresolved issue. In this study, we investigated the therapeutic effect of BMSCs overexpression of NGF on bone defect by inhibiting pyroptosis. METHODS The relationship between the low survival rate and pyroptosis of BMSCs overexpressing NGF in localized inflammation of fractures was explored by detecting pyroptosis protein levels. Then, the NGF+/BMSCs-NSA-Sca bone tissue engineering was constructed by seeding BMSCs overexpressing NGF on the allograft bone scaffold and adding the pyroptosis inhibitor necrosulfonamide(NSA). The femoral condylar defect model in the Sprague-Dawley (SD) rat was studied by micro-CT, histological, WB and PCR analyses in vitro and in vivo to evaluate the regenerative effect of bone repair. RESULTS The pyroptosis that occurs in BMSCs overexpressing NGF is associated with the nerve growth factor receptor (P75NTR) during osteogenic differentiation. Furthermore, NSA can block pyroptosis in BMSCs overexpression NGF. Notably, the analyses using the critical-size femoral condylar defect model indicated that the NGF+/BMSCs-NSA-Sca group inhibited pyroptosis significantly and had higher osteogenesis in defects. CONCLUSION NGF+/BMSCs-NSA had strong osteogenic properties in repairing bone defects. Moreover, NGF+/BMSCs-NSA-Sca mixture developed in this study opens new horizons for developing novel tissue engineering constructs.
Collapse
Affiliation(s)
- Ying Ji
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Yongkang Mao
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Honghu Lin
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Ye Wang
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Peishuai Zhao
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Yong Guo
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, 1 Zhiyuan Road, Guilin, 541199, China
| | - Lantao Gu
- Key Laboratory of Medical Biotechnology and Translational Medicine, Guilin Medical University, 1 Zhiyuan Road, Guilin, 541199, China
| | - Can Fu
- Key Laboratory of Medical Biotechnology and Translational Medicine, Guilin Medical University, 1 Zhiyuan Road, Guilin, 541199, China
| | - Ximiao Chen
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Zheng Lv
- Department of Radiology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Ning Wang
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China
| | - Qiang Li
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China.
| | - Chaoyong Bei
- Department of Orthopaedics, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, China.
| |
Collapse
|
13
|
Evrard R, Feyens M, Manon J, Lengelé B, Cartiaux O, Schubert T. Impact of NaOH based perfusion-decellularization protocol on mechanical resistance of structural bone allografts. Connect Tissue Res 2024; 65:279-292. [PMID: 38781097 DOI: 10.1080/03008207.2024.2356586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION To mitigate the post-operative complication rates associated with massive bone allografts, tissue engineering techniques have been employed to decellularize entire bones through perfusion with a sequence of solvents. Mechanical assessment was performed in order to compare conventional massive bone allografts and perfusion/decellularized massive bone allografts. MATERIAL AND METHODS Ten porcine femurs were included. Five were decellularized by perfusion. The remaining 5 were left untreated as the "control" group. Biomechanical testing was conducted on each bone, encompassing five different assessments: screw pull-out, 3-points bending, torsion, compression and Vickers indentation. RESULTS Under the experimental conditions of this study, all five destructive tested variables (maximum force until screw pull-out, maximum elongation until screw pull-out, energy to pull out the screw, fracture resistance in flexion and maximum constrain of compression) were statistically significantly superior in the control group. All seven nondestructive variables (Young's modulus in flexion, Young's modulus in shear stress, Young's modulus in compression, Elastic conventional limit in compression, lengthening to rupture in compression, resilience in compression and Vickers Hardness) showed no significant difference. DISCUSSION Descriptive statistical results suggest a tendency for the biomechanical characteristics of decellularized bone to decrease compared with the control group. However, statistical inferences demonstrated a slight significant superiority of the control group with destructive mechanical stresses. Nondestructive mechanical tests (within the elastic phase of Young's modulus) were not significantly different.
Collapse
Affiliation(s)
- Robin Evrard
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| | - Maxendre Feyens
- ECAM Brussels Engineering School, Haute Ecole ICHEC-ECAM-ISFSC, Bruxelles, Belgique
| | - Julie Manon
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
| | - Benoit Lengelé
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Plastique, Reconstructrice et Esthétique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| | - Olivier Cartiaux
- ECAM Brussels Engineering School, Haute Ecole ICHEC-ECAM-ISFSC, Bruxelles, Belgique
| | - Thomas Schubert
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| |
Collapse
|
14
|
Abaydi A, Radi J, Tbatou A, Lahrach K, Boutayb F. Tibio-Talo-Calcaneal Arthrodesis: Evaluation of the Effectiveness of a Specific Surgical Technique in 17 Cases. Cureus 2024; 16:e62014. [PMID: 38887747 PMCID: PMC11182152 DOI: 10.7759/cureus.62014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION Ankle arthrodesis is a crucial surgical intervention for advanced hindfoot conditions, aiming to restore plantigrade walking and alleviate pain. This study evaluates the effectiveness of a specific surgical approach for tibiotalocalcaneal arthrodesis (TTCA), focusing on rigorous risk factor control, corticocancellous grafting, and internal fixation using an angled retrograde femoral nail in the sagittal plane, and assesses the outcomes of this approach in terms of bone fusion and reduction of postoperative complications. MATERIALS AND METHODS This retrospective analysis includes 17 patients who underwent TTCA in a trauma-orthopedic department over seven years. Data were collected from medical records, the HOSIX software, and patient consultations. Preoperative assessments, surgical techniques, postoperative care, and follow-up evaluations were documented. RESULTS The mean age of patients was 42.4 years, with a male predominance. Surgical indications included post-traumatic arthropathy (53%), inflammatory arthropathy, ankle infectious pathologies, and Charcot foot and ankle prosthesis failures. All patients underwent standard preoperative evaluations and received corticocancellous grafts. An angled retrograde femoral nail in the sagittal plane was used for internal fixation. Postoperative immobilization lasted 6 to 8 weeks, with subsequent rehabilitation. The bone fusion rate was 100%, with a low complication rate (23.5%). DISCUSSION Our study showed a younger patient population with a male predominance, different from some previous studies. Surgical techniques, including the anterior approach combined with a lateral subtalar approach, were consistent with some studies but differed from others. Corticocancellous grafts and the angled retrograde femoral nail in the sagittal plane demonstrated favorable outcomes in terms of fusion. Complication rates were lower compared to some previous reports, highlighting potential improvements in postoperative management. CONCLUSION The surgical approach described for TTCA, emphasizing rigorous risk factor control, corticocancellous grafting, and internal fixation using an angled retrograde femoral nail in the sagittal plane, led to satisfactory bone fusion and reduced postoperative complications. These results underscore the importance of this approach in achieving optimal functional outcomes in ankle arthrodesis.
Collapse
Affiliation(s)
- Anass Abaydi
- Orthodontics, University Hospital Center Hassan II, Fès, MAR
| | - Jihad Radi
- Traumatology and Orthopedic Surgery Department A, University Hospital Center Hassan II, Fès, MAR
| | - Amine Tbatou
- Traumatology and Orthopedic Surgery Department A, University Hospital Center Hassan II, Fès, MAR
| | - Kamal Lahrach
- Traumatology and Orthopedic Surgery Department A, University Hospital Center Hassan II, Fès, MAR
| | - Fawzi Boutayb
- Traumatology and Orthopedic Surgery Department A, University Hospital Center Hassan II, Fès, MAR
| |
Collapse
|
15
|
Evrard R, Manon J, Rafferty C, Fieve L, Cornu O, Kirchgesner T, Lecouvet FE, Schubert T, Lengele B. Vascular study of decellularized porcine long bones: Characterization of a tissue engineering model. Bone 2024; 182:117073. [PMID: 38493932 DOI: 10.1016/j.bone.2024.117073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
INTRODUCTION Massive bone allografts enable the reconstruction of critical bone defects in numerous conditions (e.g. tumoral, infection or trauma). Unfortunately, their biological integration remains insufficient and the reconstruction may suffer from several postoperative complications. Perfusion-decellularization emerges as a tissue engineering potential solution to enhance osseointegration. Therefore, an intrinsic vascular study of this novel tissue engineering tool becomes essential to understand its efficacy and applicability. MATERIAL AND METHODS 32 porcine long bones (humeri and femurs) were used to assess the quality of their vascular network prior and after undergoing a perfusion-decellularization protocol. 12 paired bones were used to assess the vascular matrix prior (N = 6) and after our protocol (N = 6) by immunohistochemistry. Collagen IV, Von Willebrand factor and CD31 were targeted then quantified. The medullary macroscopic vascular network was evaluated with 12 bones: 6 were decellularized and the other 6 were, as control, not treated. All 12 underwent a contrast-agent injection through the nutrient artery prior an angio CT-scan acquisition. The images were processed and the length of medullary vessels filled with contrast agent were measured on angiographic cT images obtained in control and decellularized bones by 4 independent observers to evaluate the vascular network preservation. The microscopic cortical vascular network was evaluated on 8 bones: 4 control and 4 decellularized. After injection of gelatinous fluorochrome mixture (calcein green), non-decalcified fluoroscopic microscopy was performed in order to assess the perfusion quality of cortical vascular lacunae. RESULTS The continuity of the microscopic vascular network was assessed with Collagen IV immunohistochemistry (p-value = 0.805) while the decellularization quality was observed through CD31 and Von Willebrand factor immunohistochemistry (p-values <0.001). The macroscopic vascular network was severely impaired after perfusion-decellularization; nutrient arteries were still patent but the amount of medullary vascular channels measured was significantly higher in the control group compared to the decellularized group (p-value <0.001). On average, the observers show good agreement on these results, except in the decellularized group where more inter-observer discrepancies were observed. The microscopic vascular network was observed with green fluoroscopic signal in almost every canals and lacunae of the bone cortices, in three different bone locations (proximal metaphysis, diaphysis and distal metaphysis). CONCLUSION Despite the aggressiveness of the decellularization protocol on medullary vessels, total porcine long bones decellularized by perfusion retain an acellular cortical microvascular network. By injection through the intact nutrient arteries, this latter vascular network can still be used as a total bone infusion access for bone tissue engineering in order to enhance massive bone allografts prior implantation.
Collapse
Affiliation(s)
- R Evrard
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium.
| | - J Manon
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - C Rafferty
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, UCLouvain, Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium
| | - L Fieve
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, UCLouvain, Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium
| | - O Cornu
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - T Kirchgesner
- Département d'Imagerie Médicale, Institut de Recherche Expérimentale et Clinique (Pôle IMAG), Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - F E Lecouvet
- Département d'Imagerie Médicale, Institut de Recherche Expérimentale et Clinique (Pôle IMAG), Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - T Schubert
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - B Lengele
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, UCLouvain, Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Plastique, Reconstructrice et Esthétique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| |
Collapse
|
16
|
Aleynik DY, Zhivtscov OP, Yudin VV, Kovylin RS, Komarov RN, Charykova IN, Linkova DD, Rubtsova YP, Guseva MS, Vasyagina TI, Morozov AG, Chesnokov SA, Egorikhina MN. Specifics of Porous Polymer and Xenogeneic Matrices and of Bone Tissue Regeneration Related to Their Implantation into an Experimental Rabbit Defect. Polymers (Basel) 2024; 16:1165. [PMID: 38675083 PMCID: PMC11054212 DOI: 10.3390/polym16081165] [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: 02/13/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
This paper provides a study of two bone substitutes: a hybrid porous polymer and an osteoplastic matrix based on a bovine-derived xenograft. Both materials are porous, but their pore characteristics are different. The osteoplastic matrix has pores of 300-600 µm and the hybrid polymer has smaller pores, generally of 6-20 µm, but with some pores up to 100 µm across. SEM data confirmed the porometry results and demonstrated the different structures of the materials. Therefore, both materials were characterized by an interconnected porous structure and provided conditions for the adhesion and vital activity of human ASCs in vitro. In an experimental model of rabbit shin bone defect, it was shown that, during the 6-month observation period, neither of the materials caused negative reactions in the experimental animals. By the end of the observation period, restoration of the defects in animals in both groups was completed, and elements of both materials were preserved in the defect areas. Data from morphological examinations and CT data demonstrated that the rate of rabbit bone tissue regeneration with the hybrid polymer was comparable to that with the osteoplastic matrix. Therefore, the hybrid polymer has good potential for use in further research and improvement in biomedical applications.
Collapse
Affiliation(s)
- Diana Ya. Aleynik
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
| | - Oleg P. Zhivtscov
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
| | - Vladimir V. Yudin
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
- G. A. Razuvaev Institute of Organometallic Chemistry of Russian Academy of Sciences, Tropinina 49, Nizhny Novgorod 603950, Russia (A.G.M.)
| | - Roman S. Kovylin
- G. A. Razuvaev Institute of Organometallic Chemistry of Russian Academy of Sciences, Tropinina 49, Nizhny Novgorod 603950, Russia (A.G.M.)
| | - Roman N. Komarov
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
| | - Irina N. Charykova
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
| | - Daria D. Linkova
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
| | - Yulia P. Rubtsova
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
| | - Maria S. Guseva
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
| | - Tatyana I. Vasyagina
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
| | - Alexander G. Morozov
- G. A. Razuvaev Institute of Organometallic Chemistry of Russian Academy of Sciences, Tropinina 49, Nizhny Novgorod 603950, Russia (A.G.M.)
| | - Sergey A. Chesnokov
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
- G. A. Razuvaev Institute of Organometallic Chemistry of Russian Academy of Sciences, Tropinina 49, Nizhny Novgorod 603950, Russia (A.G.M.)
| | - Marfa N. Egorikhina
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Square 10/1, Nizhny Novgorod 603005, Russia; (D.Y.A.); (O.P.Z.); (V.V.Y.); (R.N.K.); (I.N.C.); (D.D.L.); (Y.P.R.); (M.S.G.); (T.I.V.); (S.A.C.)
| |
Collapse
|
17
|
Chen Z, Zhou X, Mo M, Hu X, Liu J, Chen L. Systematic review of the osteogenic effect of rare earth nanomaterials and the underlying mechanisms. J Nanobiotechnology 2024; 22:185. [PMID: 38627717 PMCID: PMC11020458 DOI: 10.1186/s12951-024-02442-3] [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: 02/02/2024] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
Rare earth nanomaterials (RE NMs), which are based on rare earth elements, have emerged as remarkable biomaterials for use in bone regeneration. The effects of RE NMs on osteogenesis, such as promoting the osteogenic differentiation of mesenchymal stem cells, have been investigated. However, the contributions of the properties of RE NMs to bone regeneration and their interactions with various cell types during osteogenesis have not been reviewed. Here, we review the crucial roles of the physicochemical and biological properties of RE NMs and focus on their osteogenic mechanisms. RE NMs directly promote the proliferation, adhesion, migration, and osteogenic differentiation of mesenchymal stem cells. They also increase collagen secretion and mineralization to accelerate osteogenesis. Furthermore, RE NMs inhibit osteoclast formation and regulate the immune environment by modulating macrophages and promote angiogenesis by inducing hypoxia in endothelial cells. These effects create a microenvironment that is conducive to bone formation. This review will help researchers overcome current limitations to take full advantage of the osteogenic benefits of RE NMs and will suggest a potential approach for further osteogenesis research.
Collapse
Affiliation(s)
- Ziwei Chen
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Xiaohe Zhou
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Minhua Mo
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Xiaowen Hu
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Liangjiao Chen
- Department of Orthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
18
|
Chen C, Li Z, Xu C, Kang M, Lee CS, Aghaloo T, Lee M. Self-Assembled Nanocomposite Hydrogels as Carriers for Demineralized Bone Matrix Particles and Enhanced Bone Repair. Adv Healthc Mater 2024; 13:e2303592. [PMID: 38275216 PMCID: PMC11023793 DOI: 10.1002/adhm.202303592] [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: 10/18/2023] [Revised: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Demineralized bone matrix (DBM) has been widely used as an allogeneic alternative to autologous bone graft for bone repair. However, more extensive use of DBM is limited due to its particulate nature after demineralization and rapid particle dispersion following irrigation, resulting in unpredictable osteoinductivity. Here, a new design of injectable hydrogel carriers for DBM that combine self-healing ability and osteogenic properties based on the self-assembly of guanidinylated hyaluronic acid and silica-rich nanoclays is reported. The nanoclays serve as reversible linkages to form a dynamic hydrogel network with the guanidine moieties on the polymer chains. Gelation kinetics and mechanical properties can be controlled by altering nanoclay content in the hydrogel. The resulting hydrogel exerts self-healing ability due to its dynamic crosslinks and well retains its overall performance with high DBM loading. The hydrogel exhibits great cytocompatibility and osteogenic effects mediated by the nanoclays. In vivo delivery of DBM using the nanocomposite hydrogel further demonstrates robust bone regeneration in a mouse calvarial defect model in comparison to DBM delivered with aqueous HA. This work suggests a promising hydrogel platform for many applications including therapeutic delivery and tissue engineering.
Collapse
Affiliation(s)
- Chen Chen
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Zhi Li
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Changlu Xu
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Minjee Kang
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Chung-Sung Lee
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Min Lee
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
19
|
Ingoe H, Italia K, Gilliland L, Kang HW, Karel M, Maharaj J, Cutbush K, Gupta A. The Use of Glenoid Structural Allografts for Glenoid Bone Defects in Reverse Shoulder Arthroplasty. J Clin Med 2024; 13:2008. [PMID: 38610773 PMCID: PMC11012575 DOI: 10.3390/jcm13072008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Background: The use of reverse shoulder arthroplasty as a primary and revision implant is increasing. Advances in implant design and preoperative surgical planning allow the management of complex glenoid defects. As the demand for treating severe bone loss increases, custom allograft composites are needed to match the premorbid anatomy. Baseplate composite structural allografts are used in patients with eccentric and centric defects to restore the glenoid joint line. Preserving bone stock is important in younger patients where a revision surgery is expected. The aim of this article is to present the assessment, planning, and indications of femoral head allografting for bony defects of the glenoid. Methods: The preoperative surgical planning and the surgical technique to execute the plan with a baseplate composite graft are detailed. The preliminary clinical and radiological results of 29 shoulders which have undergone this graft planning and surgical technique are discussed. Clinical outcomes included visual analogue score of pain (VAS), American Shoulder and Elbow Surgeons score (ASES), Constant-Murley score (CS), satisfaction before and after operation, and active range of motion. Radiological outcomes included graft healing and presence of osteolysis or loosening. Results: The use of composite grafts in this series has shown excellent clinical outcomes, with an overall graft complication rate in complex bone loss cases of 8%. Conclusion: Femoral head structural allografting is a valid and viable surgical option for glenoid bone defects in reverse shoulder arthroplasty.
Collapse
Affiliation(s)
- Helen Ingoe
- Queensland Unit for Advanced Shoulder Research (QUASR), Queensland University of Technology, Brisbane, QLD 4000, Australia; (H.I.); (K.I.); (L.G.); (H.W.K.); (M.K.); (J.M.); (K.C.)
| | - Kristine Italia
- Queensland Unit for Advanced Shoulder Research (QUASR), Queensland University of Technology, Brisbane, QLD 4000, Australia; (H.I.); (K.I.); (L.G.); (H.W.K.); (M.K.); (J.M.); (K.C.)
- Akunah, Brisbane, QLD 4120, Australia
| | - Luke Gilliland
- Queensland Unit for Advanced Shoulder Research (QUASR), Queensland University of Technology, Brisbane, QLD 4000, Australia; (H.I.); (K.I.); (L.G.); (H.W.K.); (M.K.); (J.M.); (K.C.)
- Akunah, Brisbane, QLD 4120, Australia
| | - Hean Wu Kang
- Queensland Unit for Advanced Shoulder Research (QUASR), Queensland University of Technology, Brisbane, QLD 4000, Australia; (H.I.); (K.I.); (L.G.); (H.W.K.); (M.K.); (J.M.); (K.C.)
| | - Mirek Karel
- Queensland Unit for Advanced Shoulder Research (QUASR), Queensland University of Technology, Brisbane, QLD 4000, Australia; (H.I.); (K.I.); (L.G.); (H.W.K.); (M.K.); (J.M.); (K.C.)
| | - Jashint Maharaj
- Queensland Unit for Advanced Shoulder Research (QUASR), Queensland University of Technology, Brisbane, QLD 4000, Australia; (H.I.); (K.I.); (L.G.); (H.W.K.); (M.K.); (J.M.); (K.C.)
| | - Kenneth Cutbush
- Queensland Unit for Advanced Shoulder Research (QUASR), Queensland University of Technology, Brisbane, QLD 4000, Australia; (H.I.); (K.I.); (L.G.); (H.W.K.); (M.K.); (J.M.); (K.C.)
- School of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| | - Ashish Gupta
- Queensland Unit for Advanced Shoulder Research (QUASR), Queensland University of Technology, Brisbane, QLD 4000, Australia; (H.I.); (K.I.); (L.G.); (H.W.K.); (M.K.); (J.M.); (K.C.)
- Akunah, Brisbane, QLD 4120, Australia
- Queensland Orthopaedic Clinic, Greenslopes Private Hospital, Brisbane, QLD 4000, Australia
| |
Collapse
|
20
|
Aboulkhair AG, AboZeid AA, Beherei HH, Kamar SS. Regenerative effect of microcarrier form of acellular dermal matrix versus bone matrix bio-scaffolds loaded with adipose stem cells on rat bone defect. Ann Anat 2024; 252:152203. [PMID: 38128745 DOI: 10.1016/j.aanat.2023.152203] [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: 07/16/2023] [Revised: 12/03/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Bone defects lead to dramatic changes in the quality of life. Acellular dermal matrix (ADM) and decellularized bone matrix (DBM) are natural scaffolds for tissue regeneration. The microcarrier scaffolds enable better vascularization and cell proliferation. This study compared the effect of microcarrier forms of DBM and ADM-loaded with adipose stem cells (ASCs) in the repair of compact bone defect in-vivo. METHODS Fifty-four male rats were divided into 4 groups; (i) Group (Gp) I: sham control; (ii) GpII: underwent femur bone defect induction and left to heal spontaneously; (iii) GpIII (ADM-Gp): included 2 subgroups; IIIa and IIIb: the bone defects were filled with non-loaded ADM and ADM-loaded with ASCs, respectively; (iv) GpIV (DBM-Gp): included 2 subgroups; IVa and IVb: the bone defects were filled with non-loaded DBM and DBM-loaded with ASCs, respectively. Animals were euthanized after 1, 2 and 3 months and their femur sections were stained with H&E, Masson's trichrome and immunohistochemistry for CD31, osteopontin and osteocalcin. RESULTS Histological analysis illustrated limited bone regeneration in the cortical defect of GpII after 3 months. The histomorphometric analysis showed significant delayed mature collagen deposition as well as CD31, osteopontin and osteocalcin expression. Superior capacity of new bone regeneration was detected with bio-scaffold micro-carriers; loaded or non-loaded with ASCs. However, DBM-loaded with ASCs displayed enhanced regeneration properties confirmed by the apparently normal architecture of the new bone and accelerated expression of CD31, osteopontin and osteocalcin in the regenerated bone after 3 months. CONCLUSIONS We concluded that decellularized scaffolds significantly improved compact bone regeneration with superiority of ASCs seeded-bone scaffolds.
Collapse
Affiliation(s)
| | - Asmaa A AboZeid
- Histology Department, Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt
| | - Hanan Hassan Beherei
- Refractories, Ceramics and Building Materials Department, National Research Centre (NRC), Giza 12622, Egypt
| | - Samaa Samir Kamar
- Histology Department, Kasr Al-ainy Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
21
|
Miszuk J, Sun H. Biomimetic Therapeutics for Bone Regeneration: A Perspective on Antiaging Strategies. Macromol Biosci 2024; 24:e2300248. [PMID: 37769439 PMCID: PMC10922069 DOI: 10.1002/mabi.202300248] [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: 05/30/2023] [Revised: 09/15/2023] [Indexed: 09/30/2023]
Abstract
Advances in modern medicine and the significant reduction in infant mortality have steadily increased the population's lifespan. As more and more people in the world grow older, incidence of chronic, noncommunicable disease is anticipated to drastically increase. Recent studies have shown that improving the health of the aging population is anticipated to provide the most cost-effective and impactful improvement in quality of life during aging-driven disease. In bone, aging is tightly linked to increased risk of fracture, and markedly decreased regenerative potential, deeming it critical to develop therapeutics to improve aging-driven bone regeneration. Biomimetics offer a cost-effective method in regenerative therapeutics for bone, where there are numerous innovations improving outcomes in young models, but adapting biomimetics to aged models is still a challenge. Chronic inflammation, accumulation of reactive oxygen species, and cellular senescence are among three of the more unique challenges facing aging-induced defect repair. This review dissects many of the innovative biomimetic approaches research groups have taken to tackle these challenges, and discusses the further uncertainties that need to be addressed to push the field further. Through these research innovations, it can be noted that biomimetic therapeutics hold great potential for the future of aging-complicated defect repair.
Collapse
Affiliation(s)
- Jacob Miszuk
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, 801 Newton Road, Iowa City, IA, 52242, United States
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, 801 Newton Road, Iowa City, IA, 52242, United States
| | - Hongli Sun
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, 801 Newton Road, Iowa City, IA, 52242, United States
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, 801 Newton Road, Iowa City, IA, 52242, United States
| |
Collapse
|
22
|
Cheng YJ, Wu TH, Tseng YS, Chen WF. Development of hybrid 3D printing approach for fabrication of high-strength hydroxyapatite bioscaffold using FDM and DLP techniques. Biofabrication 2024; 16:025003. [PMID: 38226849 DOI: 10.1088/1758-5090/ad1b20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024]
Abstract
This study develops a hybrid 3D printing approach that combines fused deposition modeling (FDM) and digital light processing (DLP) techniques for fabricating bioscaffolds, enabling rapid mass production. The FDM technique fabricates outer molds, while DLP prints struts for creating penetrating channels. By combining these components, hydroxyapatite (HA) bioscaffolds with different channel sizes (600, 800, and 1000μm) and designed porosities (10%, 12.5%, and 15%) are fabricated using the slurry casting method with centrifugal vacuum defoaming for significant densification. This innovative method produces high-strength bioscaffolds with an overall porosity of 32%-37%, featuring tightly bound HA grains and a layered surface structure, resulting in remarkable cell viability and adhesion, along with minimal degradation rates and superior calcium phosphate deposition. The HA scaffolds show hardness ranging from 1.43 to 1.87 GPa, with increasing compressive strength as the designed porosity and channel size decrease. Compared to human cancellous bone at a similar porosity range of 30%-40%, exhibiting compressive strengths of 13-70 MPa and moduli of 0.8-8 GPa, the HA scaffolds demonstrate robust strengths ranging from 40 to 73 MPa, paired with lower moduli of 0.7-1.23 GPa. These attributes make them well-suited for cancellous bone repair, effectively mitigating issues like stress shielding and bone atrophy.
Collapse
Affiliation(s)
- Yu-Jui Cheng
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Tsung-Han Wu
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Yu-Sheng Tseng
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Wen-Fan Chen
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| |
Collapse
|
23
|
Evrard R, Manon J, Maistriaux L, Rafferty C, Fieve L, Heller U, Cornu O, Gianello P, Schubert T, Lengele B. Decellularization of Massive Bone Allografts By Perfusion: A New Protocol for Tissue Engineering. Tissue Eng Part A 2024; 30:31-44. [PMID: 37698880 DOI: 10.1089/ten.tea.2023.0182] [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: 09/13/2023] Open
Abstract
In terms of large bone defect reconstructions, massive bone allografts may sometimes be the only solution. However, they are still burdened with a high postoperative complication rate. Our hypothesis is that the immunogenicity of residual cells in the graft is involved in this issue. Decellularization by perfusion might therefore be the answer to process and create more biologically effective massive bone allografts. Seventy-two porcine bones were used to characterize the efficiency of our sodium hydroxide-based decellularization protocol. A sequence of solvent perfusion through each nutrient artery was set up to ensure the complete decellularization of whole long bones. Qualitative (histology and immunohistochemistry [IHC]) and quantitative (fluoroscopic absorbance and enzyme-linked immunosorbent assay) evaluations were performed to assess the decellularization and the preservation of the extracellular matrix in the bone grafts. Cytotoxicity and compatibility were also tested. Comparatively to nontreated bones, our experiments showed a very high decellularization quality, demonstrating that perfusion is mandatory to achieve an entire decellularization. Moreover, results showed a good preservation of the bone composition and microarchitecture, Haversian systems and vascular network included. This protocol reduces the human leukocyte antigen antigenic load of the graft by >50%. The majority of measured growth factors is still present in the same amount in the decellularized bones compared to the nontreated bones. Histology and IHC show that the bones were cell compatible, noncytotoxic, and capable of inducing osteoblastic differentiation of mesenchymal stem cells. Our decellularization/perfusion protocol allowed to create decellularized long bone graft models, thanks to their inner vascular network, ready for in vivo implantation or to be further used as seeding matrices.
Collapse
Affiliation(s)
- Robin Evrard
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| | - Julie Manon
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
| | - Louis Maistriaux
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Bruxelles, Belgique
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
| | - Chiara Rafferty
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
| | - Lies Fieve
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
| | - Ugo Heller
- Centre Hospitalo-Universitaire Necker Enfants Malades, Service de Chirurgie Maxillo-Faciale et Reconstructrice, Paris, France
| | - Olivier Cornu
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| | - Pierre Gianello
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Bruxelles, Belgique
| | - Thomas Schubert
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| | - Benoit Lengele
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Plastique, Reconstructrice et Esthétique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| |
Collapse
|
24
|
Lindtner RA, Wurm A, Pirchner E, Putzer D, Arora R, Coraça-Huber DC, Schirmer M, Badzoka J, Kappacher C, Huck CW, Pallua JD. Enhancing Bone Infection Diagnosis with Raman Handheld Spectroscopy: Pathogen Discrimination and Diagnostic Potential. Int J Mol Sci 2023; 25:541. [PMID: 38203710 PMCID: PMC10778662 DOI: 10.3390/ijms25010541] [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: 11/28/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Osteomyelitis is a bone disease caused by bacteria that can damage bone. Raman handheld spectroscopy has emerged as a promising diagnostic tool for detecting bone infection and can be used intraoperatively during surgical procedures. This study involved 120 bone samples from 40 patients, with 80 samples infected with either Staphylococcus aureus or Staphylococcus epidermidis. Raman handheld spectroscopy demonstrated successful differentiation between healthy and infected bone samples and between the two types of bacterial pathogens. Raman handheld spectroscopy appears to be a promising diagnostic tool in bone infection and holds the potential to overcome many of the shortcomings of traditional diagnostic procedures. Further research, however, is required to confirm its diagnostic capabilities and consider other factors, such as the limit of pathogen detection and optimal calibration standards.
Collapse
Affiliation(s)
- Richard Andreas Lindtner
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.A.L.); (A.W.); (E.P.); (D.P.); (R.A.); (D.C.C.-H.)
| | - Alexander Wurm
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.A.L.); (A.W.); (E.P.); (D.P.); (R.A.); (D.C.C.-H.)
- Praxis Dr. Med. Univ. Alexander Wurm FA für Orthopädie und Traumatologie, Koflerweg 7, 6275 Stumm, Austria
| | - Elena Pirchner
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.A.L.); (A.W.); (E.P.); (D.P.); (R.A.); (D.C.C.-H.)
| | - David Putzer
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.A.L.); (A.W.); (E.P.); (D.P.); (R.A.); (D.C.C.-H.)
| | - Rohit Arora
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.A.L.); (A.W.); (E.P.); (D.P.); (R.A.); (D.C.C.-H.)
| | - Débora Cristina Coraça-Huber
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.A.L.); (A.W.); (E.P.); (D.P.); (R.A.); (D.C.C.-H.)
| | - Michael Schirmer
- Department of Internal Medicine, Clinic II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria;
| | - Jovan Badzoka
- Institute of Analytical Chemistry and Radiochemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (J.B.); (C.K.); (C.W.H.)
| | - Christoph Kappacher
- Institute of Analytical Chemistry and Radiochemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (J.B.); (C.K.); (C.W.H.)
| | - Christian Wolfgang Huck
- Institute of Analytical Chemistry and Radiochemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (J.B.); (C.K.); (C.W.H.)
| | - Johannes Dominikus Pallua
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.A.L.); (A.W.); (E.P.); (D.P.); (R.A.); (D.C.C.-H.)
| |
Collapse
|
25
|
Tsai YH, Chen HY, Huang TY, Chen JL, Kuo LT, Huang KC. Exploring the Role of Intraoperative Positive Culture of Allograft Bone in Subsequent Postoperative Infections among Donors and Recipients in Bone Bank Processing. Diagnostics (Basel) 2023; 14:15. [PMID: 38201323 PMCID: PMC10777897 DOI: 10.3390/diagnostics14010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Allografts have been frequently used in orthopedic procedures. The purposes of this study were to evaluate the discard rates and bacterial contamination of a bone bank, and to assess the clinical outcomes of recipients with bacterial culture-positive donor allografts. METHODS We retrospectively reviewed 1764 allografts which were harvested from living donors and stored in a bone bank from 2018 to 2022. The donors whose allografts displayed bacterial contamination at retrieval of the primary hip or knee arthroplasty were followed for microbiology and subsequent prosthetic joint infection analysis. The infected pathogens, antibiotic treatment and subsequent infection were reviewed for the intraoperative positive culture group. RESULTS The discard rate was 17%, and the bacterial contamination rate of bone retrieval was 2.15%. Thirty-eight allografts at retrieval displayed confirmed bacterial growth, and 37 patients did not reveal infective signs at 6 months follow-up. A total of 1464 allografts were stored and implanted, among which 28 allografts (1.91%) were confirmed to be positive for bacterial growth and 13 cases (0.89%) were confirmed as surgical site infections. CONCLUSIONS Our results validate the suggestion that our bone bank system performs good quality monitoring to eliminate the risk of dissemination of viral and bacterial diseases and to decrease surgical site infection after allograft implantation. By ensuring aseptic conditions and contamination-reducing strategies during harvesting and thawing, the allografts can be safely stored and implanted while limiting bacterial contamination. Our findings confirm that the intraoperative positive cultures of allografts did not contribute to subsequent postoperative surgical site infection in donors and recipients.
Collapse
Affiliation(s)
- Yao-Hung Tsai
- Department of Orthopaedic Surgery, Chia-Yi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (H.-Y.C.); (J.-L.C.); (L.-T.K.); (K.-C.H.)
- College of Medicine, Chang Gung University at Taoyuan, Taoyuan 33302, Taiwan;
| | - Hung-Yen Chen
- Department of Orthopaedic Surgery, Chia-Yi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (H.-Y.C.); (J.-L.C.); (L.-T.K.); (K.-C.H.)
| | - Tsung-Yu Huang
- College of Medicine, Chang Gung University at Taoyuan, Taoyuan 33302, Taiwan;
- Division of Infectious Diseases, Department of Internal Medicine, Chia-Yi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Jiun-Liang Chen
- Department of Orthopaedic Surgery, Chia-Yi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (H.-Y.C.); (J.-L.C.); (L.-T.K.); (K.-C.H.)
- College of Medicine, Chang Gung University at Taoyuan, Taoyuan 33302, Taiwan;
| | - Liang-Tseng Kuo
- Department of Orthopaedic Surgery, Chia-Yi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (H.-Y.C.); (J.-L.C.); (L.-T.K.); (K.-C.H.)
- College of Medicine, Chang Gung University at Taoyuan, Taoyuan 33302, Taiwan;
| | - Kuo-Chin Huang
- Department of Orthopaedic Surgery, Chia-Yi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (H.-Y.C.); (J.-L.C.); (L.-T.K.); (K.-C.H.)
- College of Medicine, Chang Gung University at Taoyuan, Taoyuan 33302, Taiwan;
| |
Collapse
|
26
|
Luo Y, Chen H, Xiu P, Zeng J, Song Y, Li T. Comparative evaluation of multi-fold rib and structural iliac bone grafts in single-segment thoracic and thoracolumbar spinal tuberculosis: clinical and radiological outcomes. J Orthop Surg Res 2023; 18:917. [PMID: 38041140 PMCID: PMC10693100 DOI: 10.1186/s13018-023-04416-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023] Open
Abstract
OBJECTIVE To compare clinical and radiological outcomes of multi-fold rib and structural iliac bone grafts, the primary autologous graft techniques in anterolateral-only surgery for single-segment thoracic and thoracolumbar spinal tuberculosis. METHODS This retrospective study included 99 patients treated from January 2014 to March 2022, categorized into 64 with multi-fold rib grafts (group A) and 35 with structural iliac bone grafts (group B). Outcomes assessed included hospital stay, operation time, intraoperative blood loss, postoperative drainage, complications, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), the Visual Analog Scale (VAS) for pain, the Oswestry Disability Index (ODI), bone fusion time, and the American Spinal Injury Association (ASIA) impairment scale grade. Segmental kyphotic angle and intervertebral height were measured radiologically before surgery and follow-up. RESULTS The mean follow-up was 63.50 ± 26.05 months for group A and 64.97 ± 26.43 months for group B (P > 0.05). All patients had achieved a clinical cure. Group A had a shorter operation time (P = 0.004). Within one week post-surgery, group B reported higher VAS scores (P < 0.0001). Neurological performance and quality of life significantly improved in both groups. No significant differences were observed in segmental kyphotic angle and intervertebral height between the groups pre- and postoperatively (P > 0.05). However, group A showed a greater segmental kyphotic angle at the final follow-up, while group B had better maintenance of kyphotic angle correction and intervertebral height (P < 0.05). Bone fusion was achieved in all patients without differences in fusion time (P > 0.05). CONCLUSIONS Multi-fold rib grafts resulted in shorter operation times and less postoperative pain, while structural iliac bone grafts provided better long-term maintenance of spinal alignment and stability, suggesting their use in cases where long-term outcomes are critical.
Collapse
Affiliation(s)
- Yuanrui Luo
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, No. 37 Guo Xue Rd, Chengdu, China
| | - Hua Chen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, No. 37 Guo Xue Rd, Chengdu, China
| | - Peng Xiu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, No. 37 Guo Xue Rd, Chengdu, China
| | - Jiancheng Zeng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, No. 37 Guo Xue Rd, Chengdu, China
| | - Yueming Song
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, No. 37 Guo Xue Rd, Chengdu, China
| | - Tao Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, No. 37 Guo Xue Rd, Chengdu, China.
| |
Collapse
|
27
|
Panahipour L, Abbasabadi AO, Wagner A, Kratochwill K, Pichler M, Gruber R. Bone Allograft Acid Lysates Change the Genetic Signature of Gingival Fibroblasts. Int J Mol Sci 2023; 24:16181. [PMID: 38003371 PMCID: PMC10671348 DOI: 10.3390/ijms242216181] [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: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Bone allografts are widely used as osteoconductive support to guide bone regrowth. Bone allografts are more than a scaffold for the immigrating cells as they maintain some bioactivity of the original bone matrix. Yet, it remains unclear how immigrating cells respond to bone allografts. To this end, we have evaluated the response of mesenchymal cells exposed to acid lysates of bone allografts (ALBA). RNAseq revealed that ALBA has a strong impact on the genetic signature of gingival fibroblasts, indicated by the increased expression of IL11, AREG, C11orf96, STC1, and GK-as confirmed by RT-PCR, and for IL11 and STC1 by immunoassays. Considering that transforming growth factor-β (TGF-β) is stored in the bone matrix and may have caused the expression changes, we performed a proteomics analysis, TGF-β immunoassay, and smad2/3 nuclear translocation. ALBA neither showed detectable TGF-β nor was the lysate able to induce smad2/3 translocation. Nevertheless, the TGF-β receptor type I kinase inhibitor SB431542 significantly decreased the expression of IL11, AREG, and C11orf96, suggesting that other agonists than TGF-β are responsible for the robust cell response. The findings suggest that IL11, AREG, and C11orf96 expression in mesenchymal cells can serve as a bioassay reflecting the bioactivity of the bone allografts.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Azarakhsh Oladzad Abbasabadi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Anja Wagner
- Core Facility Proteomics, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (K.K.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus Kratochwill
- Core Facility Proteomics, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (K.K.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
28
|
Kim M, Yoon HY. The biomechanical and biological effect of supercooling on cortical bone allograft. J Vet Sci 2023; 24:e79. [PMID: 37904641 PMCID: PMC10694378 DOI: 10.4142/jvs.23183] [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: 07/11/2023] [Revised: 09/04/2023] [Accepted: 09/13/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND The need for a storage method capable of preserving the intrinsic properties of bones without using toxic substances has always been raised. Supercooling is a relatively recently introduced preservation method that meets this need. Supercooling refers to the phenomenon of liquid in which the temperature drops below its freezing point without solidifying or crystallizing. OBJECTIVES The purpose of this study was to identify the preservation efficiency and applicability of the supercooling technique as a cortical bone allograft storage modality. METHODS The biomechanical effects of various storage methods, including deep freezing, cryopreservation, lyophilization, glycerol preservation, and supercooling, were evaluated with the three-point banding test, axial compression test, and electron microscopy. Additionally, cortical bone allografts were applied to the radial bone defect in New Zealand White rabbits to determine the biological effects. The degree of bone union was assessed with postoperative clinical signs, radiography, micro-computed tomography, and biomechanical analysis. RESULTS The biomechanical properties of cortical bone grafts preserved using glycerol and supercooling method were found to be comparable to those of normal bone while also significantly stronger than deep-frozen, cryopreserved, and lyophilized bone grafts. Preclinical research performed in rabbit radial defect models revealed that supercooled and glycerol-preserved bone allografts exhibited significantly better bone union than other groups. CONCLUSIONS Considering the biomechanical and biological superiority, the supercooling technique could be one of the optimal preservation methods for cortical bone allografts. This study will form the basis for a novel application of supercooling as a bone material preservation technique.
Collapse
Affiliation(s)
- MuYoung Kim
- Department of Small Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32611, United States of America
| | - Hun-Young Yoon
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- KU Center for Animal Blood Medical Science, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
29
|
Jurczak P, Lach S. Hydrogels as Scaffolds in Bone-Related Tissue Engineering and Regeneration. Macromol Biosci 2023; 23:e2300152. [PMID: 37276333 DOI: 10.1002/mabi.202300152] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/22/2023] [Indexed: 06/07/2023]
Abstract
Several years have passed since the medical and scientific communities leaned toward tissue engineering as the most promising field to aid bone diseases and defects resulting from degenerative conditions or trauma. Owing to their histocompatibility and non-immunogenicity, bone grafts, precisely autografts, have long been the gold standard in bone tissue therapies. However, due to issues associated with grafting, especially the surgical risks and soaring prices of the procedures, alternatives are being extensively sought and researched. Fibrous and non-fibrous materials, synthetic substitutes, or cell-based products are just a few examples of research directions explored as potential solutions. A very promising subgroup of these replacements involves hydrogels. Biomaterials resembling the bone extracellular matrix and therefore acting as 3D scaffolds, providing the appropriate mechanical support and basis for cell growth and tissue regeneration. Additional possibility of using various stimuli in the form of growth factors, cells, etc., within the hydrogel structure, extends their use as bioactive agent delivery platforms and acts in favor of their further directed development. The aim of this review is to bring the reader closer to the fascinating subject of hydrogel scaffolds and present the potential of these materials, applied in bone and cartilage tissue engineering and regeneration.
Collapse
Affiliation(s)
- Przemyslaw Jurczak
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Centre Polish Academy of Sciences, Gdansk, 80-308, Poland
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk, 80-308, Poland
| | - Slawomir Lach
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk, 80-308, Poland
| |
Collapse
|
30
|
Özger H, Alpan B, Eralp L, Valiyev N, Sungur M, Aycan OE, Salduz A. Is liquid nitrogen recycled bone and vascular fibula combination the biological reconstruction of choice in lower extremity long bone tumor-related defects? J Surg Oncol 2023; 128:902-915. [PMID: 37428092 DOI: 10.1002/jso.27385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/29/2023] [Accepted: 06/15/2023] [Indexed: 07/11/2023]
Abstract
INTRODUCTION Combination techniques, which encompass the combined use of vascularized bone grafts with massive allografts or autografts (recycled bone grafts), are especially important in the biological reconstruction of tumor-related lower extremity long bone defects. Liquid nitrogen recycled bone (frozen autograft) and free vascular fibula graft (FVFG) combination, which was coined as the "frozen hotdog (FH)" method by the authors, has not been as widely used nor its outcomes reported for significantly sized patient groups. This study aims to provide an answer to whether FH is a safe and effective reconstructive tool for limb salvage in malignant tumors of the lower extremity regarding radiological, functional, and oncological outcomes. PATIENTS AND METHODS Sixty-six (male/female: 33/33) patients, who underwent FH reconstruction for tumor-related massive defects of lower extremity long bones between 2006 and 2020, were retrospectively analyzed. The mean age was 15.8 (3.8-46.7) years. The most common tumor localizations were distal femur (42.4%) and proximal tibia (21.2%) while classic osteosarcoma and Ewing's sarcoma were the most common pathologies (60.6% and 22.7%, respectively). Mean resection and FVFG lengths were 160 (90-320) mm and 192 (125-350) mm, respectively. The mean follow-up was 73.9 (24-192) months. RESULTS The mean MSTS score was 25.4 (15-30) and the mean ISOLS radiographic score was 22.6 (13-24). Mean time to full weight bearing without any assistive devices was 15.4 (6-40) months and the median time was 12 months. MSTS score negatively correlated with resected segment length and vascular fibula length (p < 0.001; p = 0.006). Although full contact apposition of the FH segment correlated with earlier full weight bearing compared to partial apposition (mean 13.7 vs. 17.9 months) (p = 0.042), the quality of reduction did not affect the ISOLS radiographic score at LFU. Overall limb survival rate was 96.3% at 5 and 10 years while FH survival rate was 91.0% and 88.1% at 5 and 10 years. Local recurrence-free survival rates were 88.8% and 85.9%, and overall survival was 89.9% and 86.1% at 5 and 10 years, respectively. Limb length discrepancy was the most common complication with 34 (51.5%) patients while shell nonunion was seen in 21 (31.8%) patients and graft fracture in 6 (9.1%). CONCLUSION The "FH" method is a safe, effective, and extremely cost-efficient reconstructive tool for tumor-related lower extremity long bone defects. Patient compliance to protracted weight-bearing, ensuring the vitality of the FVFG, and achieving an oncologically safe resection are key factors for a successful outcome.
Collapse
Affiliation(s)
- Harzem Özger
- Department of Orthopaedics and Traumatology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Bugra Alpan
- Department of Orthopaedics and Traumatology, School of Medicine, Acıbadem Mehmet Ali Aydinlar University, Istanbul, Türkiye
| | - Levent Eralp
- Department of Orthopaedics and Traumatology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Natig Valiyev
- Department of Orthopaedics and Traumatology, Acıbadem Maslak Hospital, Istanbul, Türkiye
| | - Mustafa Sungur
- Department of Orthopaedics and Traumatology, Acıbadem Atakent Hospital, Istanbul, Türkiye
| | - Osman Emre Aycan
- Baltalimani Metin Sabanci Bone Diseases Training And Research Hospital Baltalimani Hisar Cad, Istanbul, Türkiye
| | - Ahmet Salduz
- Department of Orthopaedics and Traumatology Capa, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| |
Collapse
|
31
|
Yang J, Zhang L, Ding Q, Zhang S, Sun S, Liu W, Liu J, Han X, Ding C. Flavonoid-Loaded Biomaterials in Bone Defect Repair. Molecules 2023; 28:6888. [PMID: 37836731 PMCID: PMC10574214 DOI: 10.3390/molecules28196888] [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: 08/29/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Skeletons play an important role in the human body, and can form gaps of varying sizes once damaged. Bone defect healing involves a series of complex physiological processes and requires ideal bone defect implants to accelerate bone defect healing. Traditional grafts are often accompanied by issues such as insufficient donors and disease transmission, while some bone defect implants are made of natural and synthetic polymers, which have characteristics such as good porosity, mechanical properties, high drug loading efficiency, biocompatibility and biodegradability. However, their antibacterial, antioxidant, anti-inflammatory and bone repair promoting abilities are limited. Flavonoids are natural compounds with various biological activities, such as antitumor, anti-inflammatory and analgesic. Their good anti-inflammatory, antibacterial and antioxidant activities make them beneficial for the treatment of bone defects. Several researchers have designed different types of flavonoid-loaded polymer implants for bone defects. These implants have good biocompatibility, and they can effectively promote the expression of angiogenesis factors such as VEGF and CD31, promote angiogenesis, regulate signaling pathways such as Wnt, p38, AKT, Erk and increase the levels of osteogenesis-related factors such as Runx-2, OCN, OPN significantly to accelerate the process of bone defect healing. This article reviews the effectiveness and mechanism of biomaterials loaded with flavonoids in the treatment of bone defects. Flavonoid-loaded biomaterials can effectively promote bone defect repair, but we still need to improve the overall performance of flavonoid-loaded bone repair biomaterials to improve the bioavailability of flavonoids and provide more possibilities for bone defect repair.
Collapse
Affiliation(s)
- Jiali Yang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (J.Y.); (L.Z.); (Q.D.); (S.Z.); (S.S.); (W.L.)
- Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Lifeng Zhang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (J.Y.); (L.Z.); (Q.D.); (S.Z.); (S.S.); (W.L.)
- Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Qiteng Ding
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (J.Y.); (L.Z.); (Q.D.); (S.Z.); (S.S.); (W.L.)
| | - Shuai Zhang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (J.Y.); (L.Z.); (Q.D.); (S.Z.); (S.S.); (W.L.)
| | - Shuwen Sun
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (J.Y.); (L.Z.); (Q.D.); (S.Z.); (S.S.); (W.L.)
| | - Wencong Liu
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; (J.Y.); (L.Z.); (Q.D.); (S.Z.); (S.S.); (W.L.)
- School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543002, China
| | - Jinhui Liu
- Huashikang (Shenyang) Health Industrial Group Corporation, Shenyang 110031, China;
| | - Xiao Han
- Looking Up Starry Sky Medical Research Center, Siping 136001, China;
| | - Chuanbo Ding
- Jilin Agriculture Science and Technology College, Jilin 132101, China
| |
Collapse
|
32
|
Lindtner R, Wurm A, Kugel K, Kühn J, Putzer D, Arora R, Coraça-Huber DC, Zelger P, Schirmer M, Badzoka J, Kappacher C, Huck CW, Pallua JD. Comparison of Mid-Infrared Handheld and Benchtop Spectrometers to Detect Staphylococcus epidermidis in Bone Grafts. Bioengineering (Basel) 2023; 10:1018. [PMID: 37760120 PMCID: PMC10525239 DOI: 10.3390/bioengineering10091018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Bone analyses using mid-infrared spectroscopy are gaining popularity, especially with handheld spectrometers that enable on-site testing as long as the data quality meets standards. In order to diagnose Staphylococcus epidermidis in human bone grafts, this study was carried out to compare the effectiveness of the Agilent 4300 Handheld Fourier-transform infrared with the Perkin Elmer Spectrum 100 attenuated-total-reflectance infrared spectroscopy benchtop instrument. The study analyzed 40 non-infected and 10 infected human bone samples with Staphylococcus epidermidis, collecting reflectance data between 650 cm-1 and 4000 cm-1, with a spectral resolution of 2 cm-1 (Agilent 4300 Handheld) and 0.5 cm-1 (Perkin Elmer Spectrum 100). The acquired spectral information was used for spectral and unsupervised classification, such as a principal component analysis. Both methods yielded significant results when using the recommended settings and data analysis strategies, detecting a loss in bone quality due to the infection. MIR spectroscopy provides a valuable diagnostic tool when there is a tissue shortage and time is of the essence. However, it is essential to conduct further research with larger sample sizes to verify its pros and cons thoroughly.
Collapse
Affiliation(s)
- Richard Lindtner
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.L.); (K.K.); (J.K.); (D.P.); (R.A.); (D.C.C.-H.); (J.D.P.)
| | - Alexander Wurm
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.L.); (K.K.); (J.K.); (D.P.); (R.A.); (D.C.C.-H.); (J.D.P.)
- Praxis Dr. Med. Univ. Alexander Wurm FA für Orthopädie und Traumatologie, Koflerweg 7, 6275 Stumm, Austria
| | - Katrin Kugel
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.L.); (K.K.); (J.K.); (D.P.); (R.A.); (D.C.C.-H.); (J.D.P.)
| | - Julia Kühn
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.L.); (K.K.); (J.K.); (D.P.); (R.A.); (D.C.C.-H.); (J.D.P.)
| | - David Putzer
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.L.); (K.K.); (J.K.); (D.P.); (R.A.); (D.C.C.-H.); (J.D.P.)
| | - Rohit Arora
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.L.); (K.K.); (J.K.); (D.P.); (R.A.); (D.C.C.-H.); (J.D.P.)
| | - Débora Cristina Coraça-Huber
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.L.); (K.K.); (J.K.); (D.P.); (R.A.); (D.C.C.-H.); (J.D.P.)
| | - Philipp Zelger
- University Clinic for Hearing, Voice and Speech Disorders, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria;
| | - Michael Schirmer
- Department of Internal Medicine, Clinic II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria;
| | - Jovan Badzoka
- Institute of Analytical Chemistry and Radiochemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (J.B.); (C.K.); (C.W.H.)
| | - Christoph Kappacher
- Institute of Analytical Chemistry and Radiochemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (J.B.); (C.K.); (C.W.H.)
| | - Christian Wolfgang Huck
- Institute of Analytical Chemistry and Radiochemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (J.B.); (C.K.); (C.W.H.)
| | - Johannes Dominikus Pallua
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (R.L.); (K.K.); (J.K.); (D.P.); (R.A.); (D.C.C.-H.); (J.D.P.)
| |
Collapse
|
33
|
Huang J, Sun J, Wang Q, Mo J, Nong Y, Zhai Z, Huang X, Mo J, Lu W. A Novel Wound Therapy Modality: Autologous Wound Edge Dotted Full-Thickness Skin Grafting Improving Diabetic Foot Ulcer Healing. Int J Gen Med 2023; 16:3815-3827. [PMID: 37662508 PMCID: PMC10473433 DOI: 10.2147/ijgm.s427401] [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: 06/24/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023] Open
Abstract
Aim To explore the therapeutic efficacy of autologous wound edge-dotted full-thickness skin grafting in improving diabetic foot ulcer healing. Methods Sixty-three patients were divided into three groups: conventional wound therapy (CWT) (n = 23), platelet-rich plasma (PRP) (n = 20), and graft (n = 20). All participants were followed up for 12 weeks. The therapeutic efficacy of the three different wound treatment modalities was analyzed. Results After follow-up, 37 (58.7%) patients showed complete wound re-epithelialization, of which 10 (43.5%) occurred in the CWT group, 14 (70.0%) in the PRP group, and 13 (65.0%) in the graft group. Multivariate Cox analysis showed that the independent predictive factors for ulcer healing were different treatment modalities (graft: HR = 3.214, 95% CI=1.300-7.945, P < 0.05; platelet-rich plasma: HR = 3.075, 95% CI=1.320-7.161, P < 0.01), ABI (HR = 9.917, 95% CI=2.675-36.760, P < 0.01), and TcPO2 (HR = 1.040; 95% CI=1.005-1.076; P < 0.05). Stratified analysis showed that higher ABI in graft group or PRP group had higher wound healing rate (graft group: HR = 3.748, 95% CI=1.210-11.607, P < 0.05; PRP group: HR = 5.029, 95% CI=1.743-14.509, P < 0.05); higher TcPO2 in the graft group had higher wound healing rate (HR = 15.805, 95% CI=4.414-56.594, P < 0.01). Additionally, the wound healing time (P < 0.0167) and cumulative healing rate (P < 0.05) in both the PRP group and graft group were more advantageous. The graft group promotes wound re-epithelialization earlier and faster than in the CWT group and PRP group (P < 0.05). Meanwhile, the graft group had lower medical costs (P < 0.0167). Conclusion Autologous wound edge dotted full-thickness skin grafting has a higher cost-performance ratio than traditional diabetic foot ulcer wound care and is worthy of further clinical application.
Collapse
Affiliation(s)
- Jianhao Huang
- The Department of Endocrinology and Metabolism, Jinan University, Guangzhou, Guangdong, 510632, People’s Republic of China
- The Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Jingxia Sun
- The Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Qiu Wang
- The Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Jianming Mo
- The Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Yuechou Nong
- The Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhenwei Zhai
- The Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xiuxian Huang
- The Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Jiacheng Mo
- Information Network Center of Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Wensheng Lu
- The Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
34
|
Chang SH, Hsiao HY, Chen YH, Cheng MH, Liu JW, Huang HJ, Chou YT, Amer TAM, Vijayaraghavan P, Palanisamy S, Wang YM, Lu TT. Conjugation of bone grafts with NO-delivery dinitrosyl iron complexes promotes synergistic osteogenesis and angiogenesis in rat calvaria bone defects. J Mater Chem B 2023; 11:8007-8019. [PMID: 37530140 DOI: 10.1039/d3tb00587a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Craniofacial/jawbone deformities remain a significant clinical challenge in restoring facial/dental functions and esthetics. Despite the reported therapeutics for clinical bone tissue regeneration, the bioavailability issue of autografts and limited regeneration efficacy of xenografts/synthetic bone substitutes, however, inspire continued efforts towards functional conjugation and improvement of bioactive bone graft materials. Regarding the potential of nitric oxide (NO) in tissue engineering, herein, functional conjugation of NO-delivery dinitrosyl iron complex (DNIC) and osteoconductive bone graft materials was performed to optimize the spatiotemporal control over the delivery of NO and to activate synergistic osteogenesis and angiogenesis in rat calvaria bone defects. Among three types of biomimetic DNICs, [Fe2(μ-SCH2CH2COOH)2(NO)4] (DNIC-COOH) features a steady kinetics for cellular uptake by MC3T3-E1 osteoblast cells followed by intracellular assembly of protein-bound DNICs and release of NO. This steady kinetics for intracellular delivery of NO by DNIC-COOH rationalizes its biocompatibility and wide-spectrum cell proliferation effects on MC3T3-E1 osteoblast cells and human umbilical vein endothelial cells (HUVECs). Moreover, the bridging [SCH2CH2COOH]- thiolate ligands in DNIC-COOH facilitate its chemisorption to deproteinized bovine bone mineral (DBBM) and physisorption onto TCP (β-tricalcium phosphate), respectively, which provides a mechanism to control the kinetics for the local release of loaded DNIC-COOH. Using rats with calvaria bone defects as an in vivo model, DNIC-DBBM/DNIC-TCP promotes the osteogenic and angiogenic activity ascribed to functional conjugation of osteoconductive bone graft materials and NO-delivery DNIC-COOH. Of importance, the therapeutic efficacy of DNIC-DBBM/DNIC-TCP on enhanced compact bone formation after treatment for 4 and 12 weeks supports the potential for clinical application to regenerative medicine.
Collapse
Affiliation(s)
- Shih-Hao Chang
- Department of Periodontics, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Graduate Institute of Dental and Craniofacial Science, Chang Gung University, Taoyuan 33302, Taiwan
- Center of Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
| | - Hui-Yi Hsiao
- Center of Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Hong Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Ming-Huei Cheng
- Center of Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Jia-Wei Liu
- Center of Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Hsiao-Jo Huang
- Department of Periodontics, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Center of Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
| | - Yu-Ting Chou
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan.
| | - Tarik Abdelkareem Mostafa Amer
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan.
| | - Priya Vijayaraghavan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Sathyadevi Palanisamy
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan.
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| |
Collapse
|
35
|
Rifai A, Weerasinghe DK, Tilaye GA, Nisbet D, Hodge JM, Pasco JA, Williams LJ, Samarasinghe RM, Williams RJ. Biofabrication of functional bone tissue: defining tissue-engineered scaffolds from nature. Front Bioeng Biotechnol 2023; 11:1185841. [PMID: 37614632 PMCID: PMC10444209 DOI: 10.3389/fbioe.2023.1185841] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023] Open
Abstract
Damage to bone leads to pain and loss of movement in the musculoskeletal system. Although bone can regenerate, sometimes it is damaged beyond its innate capacity. Research interest is increasingly turning to tissue engineering (TE) processes to provide a clinical solution for bone defects. Despite the increasing biomimicry of tissue-engineered scaffolds, significant gaps remain in creating the complex bone substitutes, which include the biochemical and physical conditions required to recapitulate bone cells' natural growth, differentiation and maturation. Combining advanced biomaterials with new additive manufacturing technologies allows the development of 3D tissue, capable of forming cell aggregates and organoids based on natural and stimulated cues. Here, we provide an overview of the structure and mechanical properties of natural bone, the role of bone cells, the remodelling process, cytokines and signalling pathways, causes of bone defects and typical treatments and new TE strategies. We highlight processes of selecting biomaterials, cells and growth factors. Finally, we discuss innovative tissue-engineered models that have physiological and anatomical relevance for cancer treatments, injectable stimuli gels, and other therapeutic drug delivery systems. We also review current challenges and prospects of bone TE. Overall, this review serves as guide to understand and develop better tissue-engineered bone designs.
Collapse
Affiliation(s)
- Aaqil Rifai
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - D. Kavindi Weerasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Gebreselassie Addisu Tilaye
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - David Nisbet
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
- Laboratory of Advanced Biomaterials, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| | - Jason M. Hodge
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Julie A. Pasco
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Medicine-Western Health, The University of Melbourne, St Albans, VIC, Australia
| | - Lana J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Rasika M. Samarasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Richard J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
36
|
Hubbell PJ, Roth B, Block JE. Comparative Evaluation of Mineralized Bone Allografts for Spinal Fusion Surgery. J Funct Biomater 2023; 14:384. [PMID: 37504879 PMCID: PMC10381653 DOI: 10.3390/jfb14070384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
The primary objective of this review is to evaluate whether the degree of processing and the clinical utility of commercially available mineralized bone allografts for spine surgery meet the 2020 US Food and Drug Administration's (FDA) guideline definitions for minimal manipulation and homologous use, respectively. We also assessed the consistency of performance of these products by examining the comparative postoperative radiographic fusion rates following spine surgery. Based on the FDA's criteria for determining whether a structural allograft averts regulatory oversight and classification as a drug/device/biologic, mineralized bone allografts were judged to meet the Agency's definitional descriptions for minimal manipulation and homologous use when complying with the American Association of Tissue Banks' (AATB) accredited guidelines for bone allograft harvesting, processing, storing and transplanting. Thus, these products do not require FDA medical device clearance. Radiographic fusion rates achieved with mineralized bone allografts were uniformly high (>85%) across three published systematic reviews. Little variation was found in the fusion rates irrespective of anatomical location, allograft geometry, dimensions or indication, and in most cases, the rates were similar to those for autologous bone alone. Continued utilization of mineralized bone allografts should be encouraged across all spine surgery applications where supplemental grafts and/or segmental stability are required to support mechanically solid arthrodeses.
Collapse
Affiliation(s)
- Paul J Hubbell
- Southern Pain and Neurologic, 3939 Houma Blvd., Building 2, Suite 6, Metairie, LA 70006, USA
| | - Brandon Roth
- AZ Pain Doctors, 14420 W Meeker Blvd., Building A, Ste. 211, Sun City West, AZ 85375, USA
| | - Jon E Block
- Independent Consultant, 2210 Jackson Street, Suite 401, San Francisco, CA 94115, USA
| |
Collapse
|
37
|
Hoftiezer YAJ, Posada Alvarez C, Werenski JO, Schreuder HWB, Eberlin KR, Lee SG, Lozano-Calderón SA. Salvage of a Fractured Proximal Ulnar Osteoarticular Allograft Using a Medial Femoral Condyle Free Flap: A Case Report. JBJS Case Connect 2023; 13:01709767-202309000-00024. [PMID: 37531445 DOI: 10.2106/jbjs.cc.22.00796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
CASE We present the case of a 47-year-old paraplegic woman who underwent resection of an intermediate-grade chondrosarcoma in the proximal ulna, which was initially reconstructed with an osteoarticular allograft. However, after more than 25 years without complications, she sustained an intra-articular fracture of the allograft, which was then successfully treated using a vascularized medial femoral condyle (MFC) flap and anterolateral thigh flap. The patient has subsequently recovered her baseline elbow function, has no pain, and can use her wheelchair without restrictions. CONCLUSION Free MFC flaps are viable options to salvage osteoarticular allografts that are affected by intra-articular fractures.
Collapse
Affiliation(s)
- Yannick Albert J Hoftiezer
- Hand and Arm Center, Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts
- Department of Plastic, Reconstructive and Hand Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Carolina Posada Alvarez
- Department of Orthopaedics, University Medical Center Groningen/University of Groningen, Groningen, the Netherlands
| | - Joseph O Werenski
- Orthopaedic Oncology Service, Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - H W B Schreuder
- Department of Orthopaedic Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kyle R Eberlin
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Sang-Gil Lee
- Hand and Arm Center, Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Santiago A Lozano-Calderón
- Orthopaedic Oncology Service, Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
38
|
Krasny C, Radda C, Polke R, Schallmayer D, Borchert GH, Albrecht C. A human, allogeneic cortical bone screw for distal interphalangeal joint (DIP) arthrodesis: a retrospective cohort study with at least 10 months follow-up. Arch Orthop Trauma Surg 2023; 143:4557-4564. [PMID: 36757467 PMCID: PMC10293363 DOI: 10.1007/s00402-023-04785-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/21/2023] [Indexed: 02/10/2023]
Abstract
INTRODUCTION The prime requisites of a good digital arthrodesis are a painless and stable union in a proper position. Arthrodesis of the distal interphalangeal joint of the fingers is not without potential complications including nonunion, malunion, and deep tissue infections. The Shark Screw® is a human, cortical bone allograft for osteosynthesis and an alternative to metal or bioabsorbable devices in orthopedics and trauma surgery. The primary hypothesis is that the fusion and complication rate, using the Shark Screw®, is at least similar to those reported in the literature, using metal or bioabsorbable screws. MATERIAL AND METHODS This retrospective cohort study analyzes the fusion and complication rate and the patient satisfaction of distal interphalangeal joint arthrodesis of 27 fingers with the human allogeneic cortical bone screw. Complications, Disabilities of Arm, Shoulder, and Hand Questionnaire (Quick-DASH) score and Michigan Hand Outcomes Questionnaire (MHQ) score, grip and pinch strength and fusion angle were investigated. RESULTS The mean follow-up was 23 months. At 6 weeks after surgery, fusion was obtained for all fingers. There was no surgical complication that required revision surgery. An average fusion angle of 13.6° ± 10.7° was measured. VAS pain score decreased significantly from 6.9 before surgery to 0.14 after surgery. The Quick-DASH score decreased from 10.7 to 7.8. The MHQ score improved in all sub-scores. CONCLUSION The complication rates, using the Shark Screw® for DIP joint arthrodesis, are lower compared to the results reported in the literature for other surgical techniques. Complications related to the human allograft cortical bone screw itself were not observed. The bone screw is completely remodeled into the host bone and further hardware removal is not necessary. LEVEL OF EVIDENCE IV.
Collapse
Affiliation(s)
- Christian Krasny
- Orthopaedic Hospital Vienna-Speising, 1st Departement, Speisinger Straße 109, 1130 Vienna, Austria
| | - Christian Radda
- Orthopaedic Hospital Vienna-Speising, 1st Departement, Speisinger Straße 109, 1130 Vienna, Austria
| | - Ralf Polke
- Orthopaedic Hospital Vienna-Speising, 1st Departement, Speisinger Straße 109, 1130 Vienna, Austria
| | - Daniel Schallmayer
- Orthopaedic Hospital Vienna-Speising, 1st Departement, Speisinger Straße 109, 1130 Vienna, Austria
| | - Gudrun H. Borchert
- Dr. Borchert Medical Information Management, Egelsbacher Str. 39E, 63225 Langen, Germany
| | - Christian Albrecht
- Orthopaedic Hospital Vienna-Speising, 1st Departement, Speisinger Straße 109, 1130 Vienna, Austria
| |
Collapse
|
39
|
Fujii M, Kawano S, Ueno M, Sonohata M, Kitajima M, Tanaka S, Mawatari D, Mawatari M. Clinical results of periacetabular osteotomy with structural bone allograft for the treatment of severe hip dysplasia. Bone Joint J 2023; 105-B:743-750. [PMID: 37399069 DOI: 10.1302/0301-620x.105b7.bjj-2023-0056.r1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Aims To clarify the mid-term results of transposition osteotomy of the acetabulum (TOA), a type of spherical periacetabular osteotomy, combined with structural allograft bone grafting for severe hip dysplasia. Methods We reviewed patients with severe hip dysplasia, defined as Severin IVb or V (lateral centre-edge angle (LCEA) < 0°), who underwent TOA with a structural bone allograft between 1998 and 2019. A medical chart review was conducted to extract demographic data, complications related to the osteotomy, and modified Harris Hip Score (mHHS). Radiological parameters of hip dysplasia were measured on pre- and postoperative radiographs. The cumulative probability of TOA failure (progression to Tönnis grade 3 or conversion to total hip arthroplasty) was estimated using the Kaplan-Meier product-limited method, and a multivariate Cox proportional hazard model was used to identify predictors for failure. Results A total of 64 patients (76 hips) were included in this study. The median follow-up period was ten years (interquartile range (IQR) five to 14). The median mHHS improved from 67 (IQR 56 to 80) preoperatively to 96 (IQR 85 to 97) at the latest follow-up (p < 0.001). The radiological parameters improved postoperatively (p < 0.001), with the resulting parameters falling within the normal range in 42% to 95% of hips. The survival rate was 95% at ten years and 80% at 15 years. Preoperative Tönnis grade 2 was an independent risk factor for TOA failure. Conclusion Our findings suggest that TOA with structural bone allografting is a viable surgical option for correcting severely dysplastic acetabulum in adolescents and young adults without advanced osteoarthritis, with favourable mid-term outcomes.
Collapse
Affiliation(s)
- Masanori Fujii
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Shunsuke Kawano
- Research Center for Arthroplasty, Faculty of Medicine, Saga University, Saga, Japan
| | - Masaya Ueno
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Motoki Sonohata
- Department of Orthopaedic Surgery, JCHO Saga Central Hospital, Saga, Japan
| | - Masaru Kitajima
- Department of Orthopedic Surgery, Shiroishi Kyoritsu Hospital, Saga, Japan
| | - Shiori Tanaka
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Daisuke Mawatari
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Masaaki Mawatari
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
40
|
Goldin AN, Johnson SR, Hajdu KS, Kowalski BL, Volkmar AJ, Moran CP, Rekulapelli A, Lawrenz JM, Halpern JL, Schwartz HS, Holt GE. Surgical Fixation Method in Lower Extremity Intercalary Allograft Reconstruction After Oncologic Resection: A Comparison of Plates and Nails. J Am Acad Orthop Surg 2023; Publish Ahead of Print:00124635-990000000-00725. [PMID: 37311442 DOI: 10.5435/jaaos-d-23-00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/07/2023] [Indexed: 06/15/2023] Open
Abstract
INTRODUCTION Fixation in intercalary allograft reconstruction includes plates and intramedullary nails. The purpose of this study was to examine rates of nonunion, fracture, the overall need for revision surgery, and allograft survival based on the surgical fixation method in lower extremity intercalary allografts. METHODS A retrospective chart review was performed on 51 patients with intercalary allograft reconstruction in the lower extremity. Fixation methods compared were intramedullary fixation with nails (IMN) and extramedullary fixation with plates (EMP). Complications compared were nonunion, fracture, and wound complications. The alpha was set at 0.05 for statistical analysis. RESULTS Nonunion incidence at all allograft-to-native bone junction sites was 21% (IMN) and 25% (EMP) (P = 0.8). Fracture incidence was 24% (IMN) and 32% (EMP) (P = 0.75). Median fracture-free allograft survival was 7.9 years (IMN) and 3.2 years (EMP) (P = 0.04). Infection was seen in 18% (IMN) and 12% (EMP) (P = 0.7). The overall need for revision surgery was 59% (IMN) and 71% (EMP) (P = 0.53). Allograft survival at the final follow-up was 82% (IMN) and 65% (EMP) (P = 0.33). When the EMP group was subdivided into single plate (SP) and multiple plate (MP) groups and compared with the IMN groups, fracture rates were 24% (IMN), 8% (SP), and 48% (MP) (P = 0.04). Rates of revision surgery were 59% (IMN), 46% (SP), and 86% (MP) (P = 0.04). Allograft survival at the final follow-up was 88% (IMN), 92% (SP), and 52% (MP) (P = 0.05). DISCUSSION Median fracture-free allograft survival was notably longer for the IMN group than the EMP group; otherwise, there were no notable differences between the intramedullary and extramedullary groups. When the EMP group was subdivided into the SP and MP groups, patients with MPs had higher rates of fracture, higher rates of revision surgery, and lower overall allograft survival. LEVEL OF EVIDENCE III, Therapeutic Study, Retrospective Comparative Study.
Collapse
Affiliation(s)
- Amanda N Goldin
- From the Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN (Goldin, Johnson, Hajdu, Kowalski, Volkmar, Moran, Rekulapelli, Lawrenz, Halpern, Schwartz, and Holt), and the Department of Orthopaedics, University of California, Irvine, CA (Goldin)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Gyulay KK, Karászi P, Rédei M, Sólymos P, Schandl K, Lacza Z, Horváthy DB. Evaluation of Serum Albumin-Coated Bone Allograft for Bone Regeneration: A Seven-Year Follow-Up Study of 26 Cases. Int J Mol Sci 2023; 24:ijms24119232. [PMID: 37298182 DOI: 10.3390/ijms24119232] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
We have previously reported that serum albumin-coated bone allograft (BoneAlbumin, BA) is an effective bone substitute. It improves bone regeneration at the patellar and tibial donor sites six months after harvesting bone-patellar tendon-bone (BPTB) autografts for primary anterior cruciate ligament reconstruction (ACLR). In the present study, we examined these donor sites seven years after implantation. The study group (N = 10) received BA-enhanced autologous cancellous bone at the tibial and BA alone at the patellar site. The control group (N = 16) received autologous cancellous bone at the tibial and blood clot at the patellar site. We evaluated subcortical density, cortical thickness, and bone defect volume via CT scans. At the patellar site, subcortical density was significantly higher in the BA group at both time points. There was no significant difference in cortical thickness between the two groups at either donor site. The control group's bone defect significantly improved and reached the BA group's values at both sites by year seven. Meanwhile, the bone defects in the BA group did not change significantly and were comparable to the six-month measurements. No complications were observed. There are two limitations in this study: The number of patients recruited is small, and the randomization of the patients could have improved the quality of the study as the control group patients were older compared to the study group patients. Our 7-year results seem to demonstrate that BA is a safe and effective bone substitute that supports faster regeneration of donor sites and results in good-quality bone tissue at the time of ACLR with BPTB autografts. However, studies with a larger number of patients are required to definitively confirm the preliminary results of our study.
Collapse
Affiliation(s)
- Kata K Gyulay
- Department of Interventional Radiology, Heart and Vascular Centre, Semmelweis University, 1122 Budapest, Hungary
- Department of Radiology, Medical Imaging Centre, Semmelweis University, 1083 Budapest, Hungary
| | - Péter Karászi
- Department of Sports Surgery, Saint George University Teaching Hospital of County-Fejér, 8000 Székesfehérvár, Hungary
| | - Mátyás Rédei
- Department of Interventional Radiology, Heart and Vascular Centre, Semmelweis University, 1122 Budapest, Hungary
- Department of Radiology, Medical Imaging Centre, Semmelweis University, 1083 Budapest, Hungary
| | - Petra Sólymos
- Department of Interventional Radiology, Heart and Vascular Centre, Semmelweis University, 1122 Budapest, Hungary
- Department of Radiology, Medical Imaging Centre, Semmelweis University, 1083 Budapest, Hungary
| | - Károly Schandl
- Department of Sports Surgery, Saint George University Teaching Hospital of County-Fejér, 8000 Székesfehérvár, Hungary
| | - Zsombor Lacza
- Department of Sports Physiology, University of Physical Education, 1123 Budapest, Hungary
- Translational Medicine Institute, Semmelweis University, 1085 Budapest, Hungary
| | - Dénes B Horváthy
- Department of Interventional Radiology, Heart and Vascular Centre, Semmelweis University, 1122 Budapest, Hungary
- Department of Radiology, Medical Imaging Centre, Semmelweis University, 1083 Budapest, Hungary
| |
Collapse
|
42
|
Adeoye AO, Hadie SNH, Munajat I, Mohd Zaharri NI, Zawawi MSF, Tuan Sharif SE, Sulaiman AR. Periosteum: Functional Anatomy and Clinical Application. MALAYSIAN JOURNAL OF MEDICINE AND HEALTH SCIENCES 2023; 19:362-374. [DOI: 10.47836/mjmhs.19.3.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Periosteum is a connective tissue that envelopes the outer surface of bones and is tightly bound to the underlying bone by Sharpey’s fibers. It is composed of two layers, the outer fibrous layer and the inner cambium layer. The periosteum is densely vascularised and contains an osteoprogenitor niche that serves as a repository for bone-forming cells, which makes it an essential bone-regenerating tissue and has immensely contributed to fracture healing. Due to the high vascularity of inner cambium layer of the periosteum, periosteal transplantation has been widely used in the management of bone defects and fracture by orthopedic surgeons. Nevertheless, the use of periosteal graft in the management of bone defect is limited due to its contracted nature after being harvested. This review summarizes the current state of knowledge about the structure of periosteum, and how periosteal transplantation have been used in clinical practices, with special reference on its expansion.
Collapse
|
43
|
Ji C, Zhang C, Xu Z, Chen Y, Gan Y, Zhou M, Li L, Duan Q, Huang T, Lin J. Mussel-inspired HA@TA-CS/SA biomimetic 3D printed scaffolds with antibacterial activity for bone repair. Front Bioeng Biotechnol 2023; 11:1193605. [PMID: 37229495 PMCID: PMC10203166 DOI: 10.3389/fbioe.2023.1193605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Bacterial infection is a major challenge that could threaten the patient's life in repairing bone defects with implant materials. Developing functional scaffolds with an intelligent antibacterial function that can be used for bone repair is very important. We constructed a drug delivery (HA@TA-CS/SA) scaffold with curcumin-loaded dendritic mesoporous organic silica nanoparticles (DMON@Cur) via 3D printing for antibacterial bone repair. Inspired by the adhesion mechanism of mussels, the HA@TA-CS/SA scaffold of hydroxyapatite (HA) and chitosan (CS) is bridged by tannic acid (TA), which in turn binds sodium alginate (SA) using electrostatic interactions. The results showed that the HA@TA-CS/SA composite scaffold had better mechanical properties compared with recent literature data, reaching 68.09 MPa. It displayed excellent degradation and mineralization capabilities with strong biocompatibility in vitro. Furthermore, the antibacterial test results indicated that the curcumin-loaded scaffold inhibited S.aureus and E.coli with 99.99% and 96.56% effectiveness, respectively. These findings show that 3D printed curcumin-loaded HA@TA-CS/SA scaffold has considerable promise for bone tissue engineering.
Collapse
Affiliation(s)
- Cheng Ji
- Quanzhou Institute of Equipment Manufacturing, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Quanzhou, Fujian, China
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, China
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou, China
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, China
| | | | - Zeya Xu
- Quanzhou Institute of Equipment Manufacturing, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Quanzhou, Fujian, China
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, China
| | - Yan Chen
- Quanzhou Institute of Equipment Manufacturing, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Quanzhou, Fujian, China
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, China
| | - Yanming Gan
- Quanzhou Institute of Equipment Manufacturing, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Quanzhou, Fujian, China
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, China
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, China
| | - Minghui Zhou
- Quanzhou Institute of Equipment Manufacturing, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Quanzhou, Fujian, China
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, China
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, China
| | - Lan Li
- Quanzhou Institute of Equipment Manufacturing, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Quanzhou, Fujian, China
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, China
| | - Qinying Duan
- Quanzhou Institute of Equipment Manufacturing, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Quanzhou, Fujian, China
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, China
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, China
| | - Tingting Huang
- Quanzhou Institute of Equipment Manufacturing, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Quanzhou, Fujian, China
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, China
| | - Jinxin Lin
- Quanzhou Institute of Equipment Manufacturing, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Quanzhou, Fujian, China
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, China
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou, China
| |
Collapse
|
44
|
Visser N, Rezaie E, Ducharme A, Shin AY, Bishop AT. The effect of surgical revascularization on the mechanical properties of cryopreserved bone allograft in a porcine tibia model. J Orthop Res 2023; 41:815-822. [PMID: 35880353 DOI: 10.1002/jor.25422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/06/2022] [Accepted: 07/23/2022] [Indexed: 02/04/2023]
Abstract
Cryopreserved bone allografts(CBA) are susceptible to infection, nonunion, and late stress fracture. Although surgical revascularization by intramedullary implantation of an arteriovenous bundle (AV bundle) generates a neoangiogenic blood supply, there is potential for vascular ingrowth-mediated bone resorption to weaken the graft. For this reason, we have evaluated changes in CBA mechanical properties of structural tibial allografts with and without surgically induced angiogenesis. Cryopreserved tibia bone allografts were transplanted to reconstruct a 3.5 cm segmental tibial defect in 16 Yucatan mini pigs. Surgical revascularization was performed in half by implantation of a cranial tibial AV bundle, (revascularization group). A control group of identical size had a ligated AV bundle implanted, (ligated group). At 20 weeks micro-computed tomography (CT) measured bone mineral density (BMD) as well as bone union. Reference point indentation (RPI) compared cortex material properties, and axial compression determined the allotransplant compressive modulus. Seven of eight tibiae in the angiogenesis group were healed at both junction points at 20 weeks. Only four of eight tibiae healed in the ligated control group. There was no significant difference between the revascularization and ligated control groups in BMD and axial compression test. Similarly, RPI parameters were statistically equal. In paired comparisons with contralateral tibias, however, some RPI values were significantly worse in the ligated control group tibiae. This study demonstrates no adverse effect of surgical angiogenesis on cryopreserved structural bone allograft biomechanical properties in a large animal orthotopic segmental tibial defect model. These data suggest the potential value of surgical angiogenesis in clinical limb-sparing reconstructive surgery.
Collapse
Affiliation(s)
- Noortje Visser
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Department of Plastic and Reconstructive Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Elisa Rezaie
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Department of Plastic and Reconstructive Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Alexandra Ducharme
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander Y Shin
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Allen T Bishop
- Microvascular Research Laboratory, Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
45
|
Fadeeva IS, Teterina AY, Minaychev VV, Senotov AS, Smirnov IV, Fadeev RS, Smirnova PV, Menukhov VO, Lomovskaya YV, Akatov VS, Barinov SM, Komlev VS. Biomimetic Remineralized Three-Dimensional Collagen Bone Matrices with an Enhanced Osteostimulating Effect. Biomimetics (Basel) 2023; 8:biomimetics8010091. [PMID: 36975321 PMCID: PMC10046016 DOI: 10.3390/biomimetics8010091] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/13/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Bone grafts with a high potential for osseointegration, capable of providing a complete and effective regeneration of bone tissue, remain an urgent and unresolved issue. The presented work proposes an approach to develop composite biomimetic bone material for reconstructive surgery by deposition (remineralization) on the surface of high-purity, demineralized bone collagen matrix calcium phosphate layers. Histological and elemental analysis have shown reproduction of the bone tissue matrix architectonics, and a high-purity degree of the obtained collagen scaffolds; the cell culture and confocal microscopy have demonstrated a high biocompatibility of the materials obtained. Adsorption spectroscopy, scanning electron microscopy, microcomputed tomography (microCT) and infrared spectroscopy, and X-ray diffraction have proven the efficiency of the deposition of calcium phosphates on the surface of bone collagen scaffolds. Cell culture and confocal microscopy methods have shown high biocompatibility of both demineralized and remineralized bone matrices. In the model of heterotopic implantation in rats, at the term of seven weeks, an intensive intratrabecular infiltration of calcium phosphate precipitates, and a pronounced synthetic activity of osteoblast remodeling and rebuilding implanted materials, were revealed in remineralized bone collagen matrices in contrast to demineralized ones. Thus, remineralization of highly purified demineralized bone matrices significantly enhanced their osteostimulating ability. The data obtained are of interest for the creation of new highly effective osteoplastic materials for bone tissue regeneration and augmentation.
Collapse
Affiliation(s)
- Irina S. Fadeeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
- Correspondence: (I.S.F.); (A.Y.T.); (V.S.K.)
| | - Anastasia Yu. Teterina
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
- Correspondence: (I.S.F.); (A.Y.T.); (V.S.K.)
| | - Vladislav V. Minaychev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
| | - Anatoliy S. Senotov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Igor V. Smirnov
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
| | - Roman S. Fadeev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Polina V. Smirnova
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
| | - Vladislav O. Menukhov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Yana V. Lomovskaya
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Vladimir S. Akatov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Sergey M. Barinov
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
| | - Vladimir S. Komlev
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, Moscow 117334, Russia
- Correspondence: (I.S.F.); (A.Y.T.); (V.S.K.)
| |
Collapse
|
46
|
Kang Y, Xu J, Meng L, Su Y, Fang H, Liu J, Cheng YY, Jiang D, Nie Y, Song K. 3D bioprinting of dECM/Gel/QCS/nHAp hybrid scaffolds laden with mesenchymal stem cell-derived exosomes to improve angiogenesis and osteogenesis. Biofabrication 2023; 15. [PMID: 36756934 DOI: 10.1088/1758-5090/acb6b8] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023]
Abstract
Craniofacial bone regeneration is a coupled process of angiogenesis and osteogenesis, which, associated with infection, still remains a challenge in bone defects after trauma or tumor resection. 3D tissue engineering scaffolds with multifunctional-therapeutic properties can offer many advantages for the angiogenesis and osteogenesis of infected bone defects. Hence, in the present study, a microchannel networks-enriched 3D hybrid scaffold composed of decellularized extracellular matrix (dECM), gelatin (Gel), quaterinized chitosan (QCS) and nano-hydroxyapatite (nHAp) (dGQH) was fabricated by an extrusion 3D bioprinting technology. And enlightened by the characteristics of natural bone microstructure and the demands of vascularized bone regeneration, the exosomes (Exos) isolated from human adipose derived stem cells as angiogenic and osteogenic factors were then co-loaded into the desired dGQH20hybrid scaffold based on an electrostatic interaction. The results of the hybrid scaffolds performance characterization showed that these hybrid scaffolds exhibited an interconnected pore structure and appropriate degradability (>61% after 8 weeks of treatment), and the dGQH20hybrid scaffold displayed the highest porosity (83.93 ± 7.38%) and mechanical properties (tensile modulus: 62.68 ± 10.29 MPa, compressive modulus: 16.22 ± 3.61 MPa) among the dGQH hybrid scaffolds. Moreover, the dGQH20hybrid scaffold presented good antibacterial activities (against 94.90 ± 2.44% ofEscherichia coliand 95.41 ± 2.65% ofStaphylococcus aureus, respectively) as well as excellent hemocompatibility and biocompatibility. Furthermore, the results of applying the Exos to the dGQH20hybrid scaffold showed that the Exo promoted the cell attachment and proliferation on the scaffold, and also showed a significant increase in osteogenesis and vascularity regeneration in the dGQH@Exo scaffoldsin vitroandin vivo. Overall, this novel dECM/Gel/QCS/nHAp hybrid scaffold laden with Exo has a considerable potential application in reservation of craniofacial bone defects.
Collapse
Affiliation(s)
- Yue Kang
- Department of Breast Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, People's Republic of China.,State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Jie Xu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China.,Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, People's Republic of China
| | - Ling'ao Meng
- Department of Breast Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, People's Republic of China
| | - Ya Su
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Huan Fang
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China.,Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, People's Republic of China
| | - Jiaqi Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Daqing Jiang
- Department of Breast Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, People's Republic of China
| | - Yi Nie
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, People's Republic of China.,Key Laboratory of Green Process and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| |
Collapse
|
47
|
Rougier G, Maistriaux L, Fievé L, Xhema D, Evrard R, Manon J, Olszewski R, Szmytka F, Thurieau N, Boisson J, Kadlub N, Gianello P, Behets C, Lengelé B. Decellularized vascularized bone grafts: A preliminary in vitro porcine model for bioengineered transplantable bone shafts. Front Bioeng Biotechnol 2023; 10:1003861. [PMID: 36743653 PMCID: PMC9890275 DOI: 10.3389/fbioe.2022.1003861] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/09/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction: Durable reconstruction of critical size bone defects is still a surgical challenge despite the availability of numerous autologous and substitute bone options. In this paper, we have investigated the possibility of creating a living bone allograft, using the perfusion/decellularization/recellularization (PDR) technique, which was applied to an original model of vascularized porcine bone graft. Materials and Methods: 11 porcine bone forelimbs, including radius and ulna, were harvested along with their vasculature including the interosseous artery and then decellularized using a sequential detergent perfusion protocol. Cellular clearance, vasculature, extracellular matrix (ECM), and preservation of biomechanical properties were evaluated. The cytocompatibility and in vitro osteoinductive potential of acellular extracellular matrix were studied by static seeding of NIH-3T3 cells and porcine adipose mesenchymal stem cells (pAMSC), respectively. Results: The vascularized bone grafts were successfully decellularized, with an excellent preservation of the 3D morphology and ECM microarchitecture. Measurements of DNA and ECM components revealed complete cellular clearance and preservation of ECM's major proteins. Bone mineral density (BMD) acquisitions revealed a slight, yet non-significant, decrease after decellularization, while biomechanical testing was unmodified. Cone beam computed tomography (CBCT) acquisitions after vascular injection of barium sulphate confirmed the preservation of the vascular network throughout the whole graft. The non-toxicity of the scaffold was proven by the very low amount of residual sodium dodecyl sulfate (SDS) in the ECM and confirmed by the high live/dead ratio of fibroblasts seeded on periosteum and bone ECM-grafts after 3, 7, and 16 days of culture. Moreover, cell proliferation tests showed a significant multiplication of seeded cell populations at the same endpoints. Lastly, the differentiation study using pAMSC confirmed the ECM graft's potential to promote osteogenic differentiation. An osteoid-like deposition occurred when pAMSC were cultured on bone ECM in both proliferative and osteogenic differentiation media. Conclusion: Fully decellularized bone grafts can be obtained by perfusion decellularization, thereby preserving ECM architecture and their vascular network, while promoting cell growth and differentiation. These vascularized decellularized bone shaft allografts thus present a true potential for future in vivo reimplantation. Therefore, they may offer new perspectives for repairing large bone defects and for bone tissue engineering.
Collapse
Affiliation(s)
- Guillaume Rougier
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Department of Oncological and Cervicofacial Reconstructive Surgery, Otorhinolaryngology, Maxillofacial Surgery—Institut Curie, Paris, France
| | - Louis Maistriaux
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Pole of Experimental Surgery and Transplantation (CHEX)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,*Correspondence: Louis Maistriaux,
| | - Lies Fievé
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Daela Xhema
- Pole of Experimental Surgery and Transplantation (CHEX)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Robin Evrard
- Pole of Experimental Surgery and Transplantation (CHEX)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Neuromusculoskeletal Lab (NMSK)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Julie Manon
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Neuromusculoskeletal Lab (NMSK)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Raphael Olszewski
- Neuromusculoskeletal Lab (NMSK)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Department of Maxillofacial Surgery and Stomatology—Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Fabien Szmytka
- IMSIA, ENSTA Paris, Institut Polytechnique de Paris, Palaiseau, France
| | - Nicolas Thurieau
- IMSIA, ENSTA Paris, Institut Polytechnique de Paris, Palaiseau, France
| | - Jean Boisson
- IMSIA, ENSTA Paris, Institut Polytechnique de Paris, Palaiseau, France
| | - Natacha Kadlub
- IMSIA, ENSTA Paris, Institut Polytechnique de Paris, Palaiseau, France,Department of Maxillofacial and Reconstructive Surgery—Necker Enfants Malades, Paris, France
| | - Pierre Gianello
- Pole of Experimental Surgery and Transplantation (CHEX)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Catherine Behets
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Benoît Lengelé
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Department of Plastic and Reconstructive Surgery—Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
48
|
Muacevic A, Adler JR, Tarpada S, Kahn MD. Treatment of an Infected Tibial Shaft Non-Union Using a Novel 3D-Printed Titanium Mesh Cage: A Case Report. Cureus 2023; 15:e34212. [PMID: 36852371 PMCID: PMC9957684 DOI: 10.7759/cureus.34212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2023] [Indexed: 01/27/2023] Open
Abstract
Treating large bone defects resulting from trauma, tumors, or infection can be challenging, as current methods such as external fixation with bone transport, bone grafting, or amputation often come with high costs, high failure rates, high requirements for follow-up, and potential complications. In this case report, we present the successful treatment of a complicated, infected tibial shaft non-union by using a personalized three-dimensional (3D)-printed titanium mesh cage. This case adds to the existing body of literature by demonstrating successful integration of bone into a titanium implant and a demonstration of immediate postoperative weight bearing in the setting of suboptimal operative and psychosocial conditions. Futhermore, this report highlights the flexibility of 3D-printing technology and its ability to allow for continued limb salvage, even after a planned bone transport procedure has been interrupted. The use of 3D-printed implants customized to the patient's specific needs offers a promising new avenue for treating complex tibial pathologies, and the technology's versatility and ability to be tailored to individual patients makes it a promising tool for addressing a wide range of bone defects.
Collapse
|
49
|
Lu T, Ma N, He F, Liang Y, Ye J. Enhanced osteogenesis and angiogenesis of biphasic calcium phosphate scaffold by synergistic effect of silk fibroin coating and zinc doping. J Biomater Appl 2023; 37:1007-1017. [PMID: 36066873 DOI: 10.1177/08853282221124367] [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: 11/16/2022]
Abstract
Biphasic calcium phosphate (BCP) scaffold has been widely applied to bone regeneration because of its good biocompatibility and bone conduction property. However, the low mechanical strength and the lack of angiogenic and osteogenic induction properties have restricted its application in bone tissue regeneration. In this study, we combined zinc (Zn2+) doping and silk fibroin (SF) coating with expectation to enhance compressive strength, osteogenesis and angiogenesis of BCP scaffolds. The phase composition, morphology, porosity, compressive strength, in vitro degradation and cell behaviors were investigated systematically. Results showed that the scaffold coated with SF exhibited almost 3 times of compressive strength without compromising its porosity compared with the uncoated scaffold. Zn2+ doping and SF coating synergistically enhanced the alkaline phosphatase activity and osteogenesis-related genes expression of mouse bone mesenchymal stem cells (mBMSCs). Furthermore, SF coating notably improved the proliferation, cell viability and in vitro angiogenesis of human umbilical vein endothelial cells (HUVECs). This work provides a novel way to modify BCP scaffolds simultaneously with enhancing mechanical strength and biological properties.
Collapse
Affiliation(s)
- Teliang Lu
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, 26467South China University of Technology, Guangzhou, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, China.,Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ning Ma
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, 26467South China University of Technology, Guangzhou, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, China
| | - Fupo He
- School of Electromechanical Engineering, 47870Guangdong University of Technology, Guangzhou, China
| | - Yongyi Liang
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, 26467South China University of Technology, Guangzhou, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, China
| | - Jiandong Ye
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, 26467South China University of Technology, Guangzhou, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, China.,Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, 26467South China University of Technology, Guangzhou, China
| |
Collapse
|
50
|
Barik A, Kirtania MD. In-Vitro and In-Vivo Tracking of Cell-Biomaterial Interaction to Monitor the Process of Bone Regeneration. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|