1
|
Ji C, Zhang Z, Xu X, Song D, Zhang D. Hyperlipidemia impacts osteogenesis via lipophagy. Bone 2023; 167:116643. [PMID: 36513279 DOI: 10.1016/j.bone.2022.116643] [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: 09/17/2022] [Revised: 12/04/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022]
Abstract
The mechanism of the impact of hyperlipidemia on bone tissue homeostasis is unclear, and the role of lipophagy is yet to be investigated. This study investigated changes in lipophagy and osteogenesis levels under hyperlipemic conditions and explored the effects of lipophagy on bone regeneration. In vivo, femurs of mice with diet-induced moderate hyperlipidemia were ground out with a ball drill to create defects. In vitro, mouse osteoblast cell lines were grown in two different concentrations of the high-fat medium. We found that at hyperphysiological of lipid conditions, activation of lipophagy restored osteoblast function in a way, and similar results were observed in mice with diet-induced hyperlipidemia. Still, at suprahyperphysiological concentrations of lipid culture, the activation of lipophagy further inhibited osteogenesis, and inhibition of autophagy instead promoted osteogenesis to a small extent. These results demonstrate that lipophagy functions differently in diverse high-fat environments, suggesting that cellular and organismal changes in response to high-fat stimuli are dynamic. This may provide new ideas for improving bone dysfunction caused by lipid metabolism disorders.
Collapse
Affiliation(s)
- Chonghao Ji
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Zhanwei Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Dawei Song
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Dongjiao Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.
| |
Collapse
|
2
|
Novel Techniques and Future Perspective for Investigating Critical-Size Bone Defects. Bioengineering (Basel) 2022; 9:bioengineering9040171. [PMID: 35447731 PMCID: PMC9027954 DOI: 10.3390/bioengineering9040171] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 01/31/2023] Open
Abstract
A critical-size bone defect is a challenging clinical problem in which a gap between bone ends will not heal and will become a nonunion. The current treatment is to harvest and transplant an autologous bone graft to facilitate bone bridging. To develop less invasive but equally effective treatment options, one needs to first have a comprehensive understanding of the bone healing process. Therefore, it is imperative to leverage the most advanced technologies to elucidate the fundamental concepts of the bone healing process and develop innovative therapeutic strategies to bridge the nonunion gap. In this review, we first discuss the current animal models to study critical-size bone defects. Then, we focus on four novel analytic techniques and discuss their strengths and limitations. These four technologies are mass cytometry (CyTOF) for enhanced cellular analysis, imaging mass cytometry (IMC) for enhanced tissue special imaging, single-cell RNA sequencing (scRNA-seq) for detailed transcriptome analysis, and Luminex assays for comprehensive protein secretome analysis. With this new understanding of the healing of critical-size bone defects, novel methods of diagnosis and treatment will emerge.
Collapse
|
3
|
Abstract
PURPOSE The goal of this study was to identify bone defects of critical size in C57BL/6 mouse mandibles. MATERIALS AND METHODS Twenty-four male mice were included in this study. All mice underwent surgeries on their left mandibles. Mandibular defects of 1.0 mm (n = 8), 1.6 mm (n = 8), and 2.3 mm (n = 8) were created. For the investigation of bone healing after an 8-week period, micro-computed tomography scans and histomorphology were performed. RESULTS Mandibular bone nonunions were seen 0/8 in the 1.0-mm group, 6/8 in the 1.6-mm group, and 8/8 in the 2.3-mm group. The outcome of micro-computed tomography showed that, after 8 weeks, the bone mineral density and the bone volume to total volume ratio were significantly different among the 3 groups. The defect gaps in the nonunion 1.6- and 2.3-mm groups were filled with connective tissue, and no obvious bone formation was found. Additionally, in quantitative analysis, according to the new bone fill calculations, the percentages were 91.85% ± 8.03% in the 1.0-mm group, 59.84% ± 20.60% in the 1.6-mm group, and 15.36% ± 8.28% in the 2.3-mm group, which indicated statistically significantly lower defect healing in the 2.3-mm group. CONCLUSIONS The creation of 2.3-mm mandibular defects produces osseous nonunion in C57BL/6 mice.
Collapse
|
4
|
Tazawa R, Uchida K, Minehara H, Matsuura T, Kawamura T, Sekiguchi H, Muneshige K, Inoue S, Inoue G, Takaso M. Poly(POG)n loaded with recombinant human bone morphogenetic protein-2 accelerates new bone formation in a critical-sized bone defect mouse model. J Orthop Surg Res 2020; 15:471. [PMID: 33054796 PMCID: PMC7557057 DOI: 10.1186/s13018-020-01977-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
Background Delivery of bone morphogenetic protein-2 (BMP-2) via animal-derived absorbable collagen materials is used for the treatment of large bone defects. However, the administration of bovine proteins to humans is associated with the risk of zoonotic complications. We therefore examined the effect of combining BMP-2 with collagen-like peptides, poly(POG)n, in a critical-sized bone defect mouse model. Methods A 2-mm critical-sized bone defect was created in the femur of 9-week-old male C57/BL6J mice. Mice were randomly allocated into one of four treatment groups (n = 6 each): control (no treatment), poly(POG)n only, 0.2 μg, or 2.0 μg BMP-2 with poly(POG)n. New bone formation was monitored using soft X-ray radiographs, and bone formation at the bone defect site was examined using micro-computed tomography and histological examination at 4 weeks after surgery. Results Administration of 2.0 μg of BMP-2 with poly(POG)n promoted new bone formation and resulted in greater bone volume and bone mineral content than that observed in the control group and successfully achieved consolidation. In contrast, bone formation in all other groups was scarce. Conclusions Our findings suggest the potential of BMP-2 with poly(POG)n as a material, free from animal-derived collagen, for the treatment of large bone defects.
Collapse
Affiliation(s)
- Ryo Tazawa
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara City, Kanagawa, 252-0374, Japan. .,Shonan University of Medical Sciences Research Institute, Nishikubo 500, Chigasaki City, Kanagawa, 253-0083, Japan.
| | - Hiroaki Minehara
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Terumasa Matsuura
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Tadashi Kawamura
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Hiroyuki Sekiguchi
- Shonan University of Medical Sciences Research Institute, Nishikubo 500, Chigasaki City, Kanagawa, 253-0083, Japan
| | - Kyoko Muneshige
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Sho Inoue
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Gen Inoue
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara City, Kanagawa, 252-0374, Japan
| |
Collapse
|
5
|
Gunderson ZJ, Campbell ZR, McKinley TO, Natoli RM, Kacena MA. A comprehensive review of mouse diaphyseal femur fracture models. Injury 2020; 51:1439-1447. [PMID: 32362447 PMCID: PMC7323889 DOI: 10.1016/j.injury.2020.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 04/08/2020] [Indexed: 02/07/2023]
Abstract
Complications related to treatment of long bone fractures still stand as a major challenge for orthopaedic surgeons. Elucidation of the mechanisms of bone healing and development, and the subsequent alteration of these mechanisms to improve outcomes, typically requires animal models as an intermediary between in vitro and human clinical studies. Murine models are some of the most commonly used in translational research, and mouse fracture models are particularly diverse, offering a wide variety of customization with distinct benefits and limitations depending on the study. This review critically examines three common femur fracture models in the mouse, namely cortical hole, 3-point fracture (Einhorn), and segmental bone defect. We lay out the general procedure for execution of each model, evaluate the practical implications and important advantages/disadvantages of each and describe recent innovations. Furthermore, we explore the applications that each model is best adapted for in the context of the current state of murine orthopaedic research.
Collapse
Affiliation(s)
- Zachary J. Gunderson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Zachery R. Campbell
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Todd O. McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Roman M. Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Melissa A. Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA,Richard L. Roudebush VA Medical Center, IN, USA,Corresponding Author: Melissa A. Kacena, Ph.D., Director of Basic and Translational Research, Professor of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN 46202, (317) 278-3482 – office, (317) 278-9568 – fax
| |
Collapse
|
6
|
Ueno M, Lo CW, Barati D, Conrad B, Lin T, Kohno Y, Utsunomiya T, Zhang N, Maruyama M, Rhee C, Huang E, Romero-Lopez M, Tong X, Yao Z, Zwingenberger S, Yang F, Goodman SB. Interleukin-4 overexpressing mesenchymal stem cells within gelatin-based microribbon hydrogels enhance bone healing in a murine long bone critical-size defect model. J Biomed Mater Res A 2020; 108:2240-2250. [PMID: 32363683 DOI: 10.1002/jbm.a.36982] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/21/2020] [Accepted: 03/28/2020] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cell (MSC)-based therapy is a promising strategy for bone repair. Furthermore, the innate immune system, and specifically macrophages, plays a crucial role in the differentiation and activation of MSCs. The anti-inflammatory cytokine Interleukin-4 (IL-4) converts pro-inflammatory M1 macrophages into a tissue regenerative M2 phenotype, which enhances MSC differentiation and function. We developed lentivirus-transduced IL-4 overexpressing MSCs (IL-4 MSCs) that continuously produce IL-4 and polarize macrophages toward an M2 phenotype. In the current study, we investigated the potential of IL-4 MSCs delivered using a macroporous gelatin-based microribbon (μRB) scaffold for healing of critical-size long bone defects in Mice. IL-4 MSCs within μRBs enhanced M2 marker expression without inhibiting M1 marker expression in the early phase, and increased macrophage migration into the scaffold. Six weeks after establishing the bone defect, IL-4 MSCs within μRBs enhanced bone formation and helped bridge the long bone defect. IL-4 MSCs delivered using macroporous μRB scaffold is potentially a valuable strategy for the treatment of critical-size long bone defects.
Collapse
Affiliation(s)
- Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Chi-Wen Lo
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Danial Barati
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Bogdan Conrad
- Stem Cell Biology and Regenerative Medicine Program, Stanford University, Stanford, California, USA
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yusuke Kohno
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Ejun Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Monica Romero-Lopez
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Xinming Tong
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Stefan Zwingenberger
- University Center for Orthopaedics and Traumatology, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA.,Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA.,Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
7
|
Effects of Strontium-Doped β-Tricalcium Scaffold on Longitudinal Nuclear Factor-Kappa Beta and Vascular Endothelial Growth Factor Receptor-2 Promoter Activities during Healing in a Murine Critical-Size Bone Defect Model. Int J Mol Sci 2020; 21:ijms21093208. [PMID: 32370039 PMCID: PMC7246816 DOI: 10.3390/ijms21093208] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023] Open
Abstract
It was hypothesized that strontium (Sr)-doped β-tricalcium phosphate (TCP)-based scaffolds have a positive effect on the regeneration of large bone defects (LBD). Readouts in our mice models were nuclear factor-kappa beta (NF-κB) activity and vascular endothelial growth factor receptor-2 (VEGFR-2) promoter activity during the healing process. A 2-mm critical-size femoral fracture was performed in transgenic NF-κB- and VEGFR-2-luciferase reporter mice. The fracture was filled with a 3D-printed β-TCP scaffold with or without Sr. A bioluminescence in-vivo imaging system was used to sequentially investigate NF-κB and VEGFR-2 expression for two months. After sacrifice, soft and osseous tissue formation in the fracture sites was histologically examined. NF-κB activity increased in the β-TCP + Sr group in the latter stage (day 40–60). VEGFR-2 activity increased in the + Sr group from days 0–15 but decreased and showed significantly less activity than the β-TCP and non-scaffold groups from days 40–60. The new bone formation and soft tissue formation in the + Sr group were significantly higher than in the β-TCP group, whereas the percentage of osseous tissue formation in the β-TCP group was significantly higher than in the β-TCP + Sr group. We analyzed longitudinal VEGFR-2 promoter activity and NF-κB activity profiles, as respective agents of angiogenesis and inflammation, during LBD healing. The extended inflammation phase and eventually more rapid resorption of scaffold caused by the addition of strontium accelerates temporary bridging of the fracture gaps. This finding has the potential to inform an improved treatment strategy for patients who suffer from osteoporosis.
Collapse
|
8
|
Effect of Single Injection of Recombinant Human Bone Morphogenetic Protein-2-Loaded Artificial Collagen-Like Peptide in a Mouse Segmental Bone Transport Model. BIOMED RESEARCH INTERNATIONAL 2020; 2019:1014594. [PMID: 31950029 PMCID: PMC6948306 DOI: 10.1155/2019/1014594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/09/2019] [Indexed: 11/17/2022]
Abstract
This study aimed to investigate whether a single injection of recombinant human bone morphogenetic protein-2-loaded artificial collagen-like peptide gel (rhBMP-2/ACG) accelerates consolidation at the bone defect site and bone union at the docking site in a mouse segmental bone transport (SBT) model. A critical sized bone defect (2 mm) was created in the femur of mice and subsequently reconstructed using SBT with an external fixator. Mice were divided into four treatment groups: Group CONT (immobile control), Group 0.2 (bone segments moved 0.2 mm/day for 10 days), Group 1.0 (bone segments moved 1.0 mm/day for 2 days), and Group 1.0/BMP-2 (rhBMP-2/ACG injected into the bone defect and segments moved 1.0 mm/day for 2 days). Consolidation at the bone defect site and bone union at the docking site was evaluated radiologically and histologically across eight weeks. Bone volume and bone mineral content were significantly higher in Group 0.2 than in Group 1.0. Group 0.2 showed evidence of rebuilding of the medullary canal eight weeks after surgery at the bone defect site. However, in Group 1.0, maturation of regenerative bone at the bone defect site was poor, with the central area between the proximal and distal bone composed mainly of masses of fibrous and adipose tissue. Group 1.0/BMP-2 had higher bone volume and bone mineral content compared to Group 1.0, and all mice achieved bone union at the bone defect and docking sites. Single injection of rhBMP-2/ACG combined with SBT may be effective for enhancing bone healing in large bone defects.
Collapse
|
9
|
Taschieri S, Lolato A, Ofer M, Testori T, Francetti L, Del Fabbro M. Immediate post-extraction implants with or without pure platelet-rich plasma: a 5-year follow-up study. Oral Maxillofac Surg 2017; 21:147-157. [PMID: 28168420 DOI: 10.1007/s10006-017-0609-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/30/2017] [Indexed: 06/06/2023]
Abstract
PURPOSE The purpose of this study was comparison of clinical and radiographic outcomes of immediate post-extraction implants with or without the use of pure platelet-rich plasma (P-PRP) in the short- and medium-term follow-up. METHODS A retrospective analysis was performed to assess soft tissue healing, implant and prosthesis survival, marginal bone level changes and biological complications. RESULTS A total of 109 partially edentulous patients with 126 implants were included in this analysis. At 4-5 years after loading, cumulative survival rate in test group was 97.4% and in control group was 97.8%, with no significant differences. After 5 years of function, marginal bone loss (MBL) in test group was 0.8 ± 0.35 and 1.02 ± 0.27 mm for immediate and delayed loading, respectively, and in control group was 0.6 ± 0.16 and 0.8 ± 0.89 mm for immediate and delayed loading, respectively. No significant differences in MBL were observed intragroups and intergroups at any time point considered. Soft tissue healing score was significantly higher in test group compared to the control at 3 and 7 days after surgery, with significant differences. CONCLUSIONS P-PRP implant group showed a better soft tissue management and wound healing in the first 7 days after surgery compared to non-P-PRP implant group. In the medium-term follow-up, comparable clinical and radiographic outcomes were noticed between two groups.
Collapse
Affiliation(s)
- Silvio Taschieri
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Alessandra Lolato
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Moses Ofer
- Department of Periodontology & Dental Implantology, School of Dental Medicine, University of Tel Aviv, Tel Aviv-Yafo, Israel
| | - Tiziano Testori
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Luca Francetti
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Massimo Del Fabbro
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy.
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.
| |
Collapse
|
10
|
Manassero M, Decambron A, Huu Thong BT, Viateau V, Bensidhoum M, Petite H. Establishment of a Segmental Femoral Critical-size Defect Model in Mice Stabilized by Plate Osteosynthesis. J Vis Exp 2016. [PMID: 27768070 PMCID: PMC5092194 DOI: 10.3791/52940] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The use of tissue-engineered bone constructs is an appealing strategy to overcome drawbacks of autografts for the treatment of massive bone defects. As a model organism, the mouse has already been widely used in bone-related research. Large diaphyseal bone defect models in mice, however, are sparse and often use bone fixation which fills the bone marrow cavity and does not provide optimal mechanical stability. The objectives of the current study were to develop a critical-size, segmental, femoral defect in nude mice. A 3.5-mm mid-diaphyseal femoral ostectomy (approximately 25% of the femur length) was performed using a dedicated jig, and was stabilized with an anterior located locking plate and 4 locking screws. The bone defect was subsequently either left empty or filled with a bone substitute (syngenic bone graft or coralline scaffold). Bone healing was monitored noninvasively using radiography and in vivo micro-computed-tomography and was subsequently assessed by ex vivo micro-computed-tomography and undecalcified histology after animal sacrifice, 10 weeks postoperatively. The recovery of all mice was excellent, a full-weight-bearing was observed within one day following the surgical procedure. Furthermore, stable bone fixation and consistent fixation of the implanted materials were achieved in all animals tested throughout the study. When the bone defects were left empty, non-union was consistently obtained. In contrast, when the bone defects were filled with syngenic bone grafts, bone union was always observed. When the bone defects were filled with coralline scaffolds, newly-formed bone was observed in the interface between bone resection edges and the scaffold, as well as within a short distance within the scaffold. The present model describes a reproducible critical-size femoral defect stabilized by plate osteosynthesis with low morbidity in mice. The new load-bearing segmental bone defect model could be useful for studying the underlying mechanisms in bone regeneration pertinent to orthopaedic applications.
Collapse
Affiliation(s)
- Mathieu Manassero
- Laboratoire de Bioingénierie et Biomécanique Ostéo-Articulaires (B2OA - UMR CNRS 7052), Université Paris Diderot; Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est
| | - Adeline Decambron
- Laboratoire de Bioingénierie et Biomécanique Ostéo-Articulaires (B2OA - UMR CNRS 7052), Université Paris Diderot; Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est
| | - Bui Truong Huu Thong
- Laboratoire de Bioingénierie et Biomécanique Ostéo-Articulaires (B2OA - UMR CNRS 7052), Université Paris Diderot
| | - Véronique Viateau
- Laboratoire de Bioingénierie et Biomécanique Ostéo-Articulaires (B2OA - UMR CNRS 7052), Université Paris Diderot; Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est
| | - Morad Bensidhoum
- Laboratoire de Bioingénierie et Biomécanique Ostéo-Articulaires (B2OA - UMR CNRS 7052), Université Paris Diderot
| | - Hervé Petite
- Laboratoire de Bioingénierie et Biomécanique Ostéo-Articulaires (B2OA - UMR CNRS 7052), Université Paris Diderot;
| |
Collapse
|
11
|
Carlier A, Skvortsov GA, Hafezi F, Ferraris E, Patterson J, Koç B, Van Oosterwyck H. Computational model-informed design and bioprinting of cell-patterned constructs for bone tissue engineering. Biofabrication 2016; 8:025009. [DOI: 10.1088/1758-5090/8/2/025009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
12
|
Falah M, Rayan A, Srouji S. Storage effect on viability and biofunctionality of human adipose tissue-derived stromal cells. Cytotherapy 2016; 17:1220-9. [PMID: 26276005 DOI: 10.1016/j.jcyt.2015.05.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 05/05/2015] [Accepted: 05/26/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND AIMS In our recent studies, the transplantation of human adipose tissue-derived stromal cells (ASCs) has shown promise for treatment of diseases related to bone and joint disorders. METHODS For the current clinical applications, ASCs were formulated and suspended in PlasmaLyte A supplemented with heparin, glucose and human serum albumin, balanced to pH 7.4 with sodium bicarbonate. This cell solution constitutes 20% of the overall transplanted mixture and is supplemented with hyaluronic acid (60%) and OraGraft particles (20%). We intended to investigate the effect of this transplantation mixture on the viability and biofunctionality of ASCs in bone formation. Freshly harvested cells were resuspended and incubated in the indicated mixture for up to 48 h at 4°C. Cell viability was assessed using trypan blue and AlamarBlue, and cell functionality was determined by quantifying their adhesion rate in vitro and bone formation in an ectopic mouse model. RESULTS More than 80% of the ASCs stored in the transplantation mixture were viable for up to 24 h. Cell viability beyond 24 h in storage decreased to approximately 50%. In addition, an equal degree of bone formation was observed between the cells transplanted following incubation in transplantation mixture for up to 24 h and zero-time non-incubated cells (control). CONCLUSIONS The viability and functionality of ASCs stored in the presented formulation will make such cell therapy accessible to larger and more remote populations.
Collapse
Affiliation(s)
- Mizied Falah
- Drug Discovery Informatics Lab, QRC-Qasemi Research Center, Al-Qasemi Academic College, Baka El-Garbiah, Israel
| | - Anwar Rayan
- Drug Discovery Informatics Lab, QRC-Qasemi Research Center, Al-Qasemi Academic College, Baka El-Garbiah, Israel
| | - Samer Srouji
- Oral and Maxillofacial Surgery Department, Bone Regeneration Lab, Galilee Medical Center, Nahariya, Israel; Faculty of Medicine in the Galilee, Bar-ilan University, Ramat Gan, Israel.
| |
Collapse
|
13
|
Carlier A, Lammens J, Van Oosterwyck H, Geris L. Computational modeling of bone fracture non-unions: four clinically relevant case studies. IN SILICO CELL AND TISSUE SCIENCE 2015; 2:1. [PMID: 26709368 PMCID: PMC4684906 DOI: 10.1186/s40482-015-0004-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/11/2015] [Indexed: 12/02/2022]
Abstract
The human skeleton has a remarkable regeneration capacity. Nevertheless, 5-10 % of the bone fractures fails to heal and develops into a non-union which is a challenging orthopedic complication requiring complex and expensive treatment. This review paper will discuss four different computational models, each capturing a particular clinical case of non-union: non-union induced by reaming of the marrow canal and periosteal stripping, non-union due to a large interfragmentary gap, non-union due to a genetic disorder [i.e. NF1 related congenital pseudoarthrosis of the tibia (CPT)] and non-union due to mechanical overload. Together, the four computational models are able to capture the etiology of a wide range of fracture non-union types and design novel treatment strategies thereof. Further research is required to corroborate the computational models in both animal and human settings and translate them from bench to bed side.
Collapse
Affiliation(s)
- Aurélie Carlier
- />Biomechanics Section, KU Leuven, Celestijnenlaan 300 C, PB 2419, 3000 Leuven, Belgium
- />Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Johan Lammens
- />Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- />Department of Orthopaedics, University Hospitals of KU Leuven, KU Leuven, Weligerveld 1-blok 1, 3212 Pellenberg, Belgium
| | - Hans Van Oosterwyck
- />Biomechanics Section, KU Leuven, Celestijnenlaan 300 C, PB 2419, 3000 Leuven, Belgium
- />Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Liesbet Geris
- />Biomechanics Section, KU Leuven, Celestijnenlaan 300 C, PB 2419, 3000 Leuven, Belgium
- />Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- />Biomechanics Research Unit, Department of Aerospace and Mechanical Engineering, University of Liege, Chemin des Chevreuils 1-BAT 52/3, 4000 Liege 1, Belgium
| |
Collapse
|
14
|
Cignachi NP, Pesquero JB, Oliveira RB, Etges A, Campos MM. Kinin B1Receptor Deletion Affects Bone Healing in Type 1 Diabetic Mice. J Cell Physiol 2015; 230:3019-28. [DOI: 10.1002/jcp.25034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 05/05/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Natália P. Cignachi
- School of Dentistry; Pontifical Catholic University of Rio Grande do Sul; Avenida Ipiranga; 6681; Partenon; Porto Alegre RS Brazil
| | - João B. Pesquero
- Department of Biophysics; Federal University of São Paulo; Rua Pedro de Toledo, 669; São Paulo SP Brazil
| | - Rogério B. Oliveira
- School of Dentistry; Pontifical Catholic University of Rio Grande do Sul; Avenida Ipiranga; 6681; Partenon; Porto Alegre RS Brazil
| | - Adriana Etges
- Department of Oral Pathology; School of Dentistry; Universidade Federal de Pelotas (UFPel); Rua, Gonçalves Chaves, 457; Pelotas RS Brazil
| | - Maria M. Campos
- School of Dentistry; Pontifical Catholic University of Rio Grande do Sul; Avenida Ipiranga; 6681; Partenon; Porto Alegre RS Brazil
- Institute of Toxicology and Pharmacology; Pontifical Catholic University of Rio Grande do Sul; Avenida Ipiranga, 6681; Partenon; Porto Alegre RS Brazil
| |
Collapse
|
15
|
Carlier A, van Gastel N, Geris L, Carmeliet G, Van Oosterwyck H. Size does matter: an integrative in vivo-in silico approach for the treatment of critical size bone defects. PLoS Comput Biol 2014; 10:e1003888. [PMID: 25375821 PMCID: PMC4222588 DOI: 10.1371/journal.pcbi.1003888] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 09/02/2014] [Indexed: 01/07/2023] Open
Abstract
Although bone has a unique restorative capacity, i.e., it has the potential to heal scarlessly, the conditions for spontaneous bone healing are not always present, leading to a delayed union or a non-union. In this work, we use an integrative in vivo - in silico approach to investigate the occurrence of non-unions, as well as to design possible treatment strategies thereof. The gap size of the domain geometry of a previously published mathematical model was enlarged in order to study the complex interplay of blood vessel formation, oxygen supply, growth factors and cell proliferation on the final healing outcome in large bone defects. The multiscale oxygen model was not only able to capture the essential aspects of in vivo non-unions, it also assisted in understanding the underlying mechanisms of action, i.e., the delayed vascularization of the central callus region resulted in harsh hypoxic conditions, cell death and finally disrupted bone healing. Inspired by the importance of a timely vascularization, as well as by the limited biological potential of the fracture hematoma, the influence of the host environment on the bone healing process in critical size defects was explored further. Moreover, dependent on the host environment, several treatment strategies were designed and tested for effectiveness. A qualitative correspondence between the predicted outcomes of certain treatment strategies and experimental observations was obtained, clearly illustrating the model's potential. In conclusion, the results of this study demonstrate that due to the complex non-linear dynamics of blood vessel formation, oxygen supply, growth factor production and cell proliferation and the interactions thereof with the host environment, an integrative in silico-in vivo approach is a crucial tool to further unravel the occurrence and treatments of challenging critical sized bone defects. In 5–10% of fracture patients, the bone fractures do not heal in the normal healing period (delayed healing) or do not heal at all (non-union). In order to investigate the causes of impaired healing and design potential treatment strategies, we have used a combined experimental and computational approach. More specifically, large bone defects were analyzed in mouse models and simulated by a previously published computational model. After showing that the predictions of the computational model match the observations of the experimental model, we have used the computational model to investigate the underlying mechanisms of action. In particular, the results indicated that the new blood vessels do not reach the central fracture zone in time due to the large defect size, which leads to insufficient oxygen delivery, increased cell death and disrupted bone healing. The healing, however, could be rescued by adequate blood vessel ingrowth from the overlying soft tissues. Moreover, potential treatment strategies were designed based on the influence of these soft tissues. In conclusion, this study demonstrates the potential of a combined experimental and computational approach to contribute to the understanding of pathological processes like the impaired bone regeneration in large bone defects and design future treatments thereof.
Collapse
Affiliation(s)
- Aurélie Carlier
- Biomechanics Section, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Biomechanics Research Unit, University of Liège, Liège, Belgium
- * E-mail:
| | - Nick van Gastel
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Biomechanics Section, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Biomechanics Research Unit, University of Liège, Liège, Belgium
| | - Geert Carmeliet
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Hans Van Oosterwyck
- Biomechanics Section, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Viateau V, Decambron A, Manassero M. Animal Models for Orthopedic Applications of Tissue Engineering. Biomaterials 2014. [DOI: 10.1002/9781119043553.ch8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
17
|
Xing J, Jin H, Hou T, Chang Z, Luo F, Wang P, Li Z, Xie Z, Xu J. Establishment of a bilateral femoral large segmental bone defect mouse model potentially applicable to basic research in bone tissue engineering. J Surg Res 2014; 192:454-63. [PMID: 24972741 DOI: 10.1016/j.jss.2014.05.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 01/17/2014] [Accepted: 05/16/2014] [Indexed: 12/01/2022]
Abstract
BACKGROUND To understand the cellular mechanism underlying bone defect healing in the context of tissue engineering, a reliable, reproducible, and standardized load-bearing large segmental bone defect model in small animals is indispensable. The aim of this study was to establish and evaluate a bilateral femoral defect model in mice. MATERIALS AND METHODS Donor mouse bone marrow mesenchymal stem cells (mBMSCs) were obtained from six mice (FVB/N) and incorporated into partially demineralized bone matrix scaffolds to construct tissue-engineered bones. In total, 36 GFP(+) mice were used for modeling. Titanium fixation plates with locking steel wires were attached to the femurs for stabilization, and 2-mm-long segmental bone defects were created in the bilateral femoral midshafts. The defects in the left and right femurs were transplanted with tissue-engineered bones and control scaffolds, respectively. The healing process was monitored by x-ray radiography, microcomputed tomography, and histology. The capacity of the transplanted mBMSCs to recruit host CD31(+) cells was investigated by immunofluorescence and real-time polymerase chain reaction. RESULTS Postoperatively, no complication was observed, except that two mice died of unknown causes. Stable fixation of femurs and implants with full load bearing was achieved in all animals. The process of bone defect repair was significantly accelerated due to the introduction of mBMSCs. Moreover, the transplanted mBMSCs attracted more host CD31(+) endothelial progenitors into the grafts. CONCLUSIONS The present study established a feasible, reproducible, and clinically relevant bilateral femoral large segmental bone defect mouse model. This model is potentially suitable for basic research in the field of bone tissue engineering.
Collapse
Affiliation(s)
- Junchao Xing
- National and Local United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, the Third Military Medical University, Chongqing, China; Laboratory of Tissue Engineering in Chongqing City, Chongqing, 400038, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, 400038, China
| | - Huiyong Jin
- National and Local United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, the Third Military Medical University, Chongqing, China; Laboratory of Tissue Engineering in Chongqing City, Chongqing, 400038, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, 400038, China; Department of Orthopaedics, No. 519 Hospital of PLA, Xichang, 615000, China
| | - Tianyong Hou
- National and Local United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, the Third Military Medical University, Chongqing, China; Laboratory of Tissue Engineering in Chongqing City, Chongqing, 400038, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, 400038, China.
| | - Zhengqi Chang
- National and Local United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, the Third Military Medical University, Chongqing, China; Laboratory of Tissue Engineering in Chongqing City, Chongqing, 400038, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, 400038, China
| | - Fei Luo
- National and Local United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, the Third Military Medical University, Chongqing, China; Laboratory of Tissue Engineering in Chongqing City, Chongqing, 400038, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, 400038, China
| | - Pinpin Wang
- National and Local United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, the Third Military Medical University, Chongqing, China; Laboratory of Tissue Engineering in Chongqing City, Chongqing, 400038, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, 400038, China
| | - Zhiqiang Li
- National and Local United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, the Third Military Medical University, Chongqing, China; Laboratory of Tissue Engineering in Chongqing City, Chongqing, 400038, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, 400038, China
| | - Zhao Xie
- National and Local United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, the Third Military Medical University, Chongqing, China; Laboratory of Tissue Engineering in Chongqing City, Chongqing, 400038, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, 400038, China
| | - Jianzhong Xu
- National and Local United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, the Third Military Medical University, Chongqing, China; Laboratory of Tissue Engineering in Chongqing City, Chongqing, 400038, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, 400038, China.
| |
Collapse
|
18
|
Liu K, Li D, Huang X, Lv K, Ongodia D, Zhu L, Zhou L, Li Z. A murine femoral segmental defect model for bone tissue engineering using a novel rigid internal fixation system. J Surg Res 2013; 183:493-502. [PMID: 23522461 DOI: 10.1016/j.jss.2013.02.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/04/2013] [Accepted: 02/20/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND As a model animal, the mouse has already been widely used in bone-related research. However, there is a lack of ideal long bone segmental defect mouse model. Since external fixation has disadvantages of heavy weight, penetrating the skin, and hampering mobility, an internal fixation device is probably more preferable to maintain the segmental bone defect. The aim of this study was to establish a simple, reproducible, and standardized murine critical-size defect model through designing an internal fixation system, verifying its adaptability, and investigating the critical size of femoral segmental defect. METHODS By utilizing computer-aided measuring and processing system, anatomical data of adult C57BL/6 mouse femur was obtained, and a plate-bolts system was designed for rigid fixation. The plate and screws were fixed in 67 mice and 1.5 or 2.0 mm defect gaps were created in the femoral midshaft. Compression and three-point bending of bone-implant construct were tested in mice at 0, 2, 5, and 12 wk postoperative to test the biomechanical stability. X-ray, micro-computed tomography, and histology were used to investigate the defect healing process. RESULTS The plate- and screws-fitted mouse femur and unilateral or bilateral operation had seemingly no adverse impact on the mouse in general. Mechanical tests indicated that there were no significant differences between the bone-implant construct and intact femur in compression and three-point bending loading. Micro-computed tomography scanning showed the bone mineral density had not been affected by the implantation of fixation device. There was no union of the 2.0 mm segmental defect in 12-wk period. CONCLUSION Using the specifically designed rigid internal fixation device, a segmental defect size of 2.0 mm in C57BL/6 mouse femur will show nonunion and can serve as a critical defect size for bone tissue engineering and bone regeneration research.
Collapse
Affiliation(s)
- Kai Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Montijo HE, Kellam JF, Gettys FK, Starman JS, Nelson MAJKJ, Bayoumi EM, Bosse MJ, Gruber HE. Utilization of the AO LockingRatNail in a Novel Rat Femur Critical Defect Model. J INVEST SURG 2012; 25:381-6. [DOI: 10.3109/08941939.2012.655370] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
20
|
Manassero M, Viateau V, Matthys R, Deschepper M, Vallefuoco R, Bensidhoum M, Petite H. A novel murine femoral segmental critical-sized defect model stabilized by plate osteosynthesis for bone tissue engineering purposes. Tissue Eng Part C Methods 2012; 19:271-80. [PMID: 22953787 DOI: 10.1089/ten.tec.2012.0256] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mouse models are invaluable tools for mechanistic and efficacy studies of the healing process of large bone defects resulting in atrophic nonunions, a severe medical problem and a financial health-care-related burden. Models of atrophic nonunions are usually achieved by providing a highly stable biomechanical environment. For this purpose, external fixators have been investigated, but plate osteosynthesis, despite its high clinical relevance, has not yet been considered in mice. We hereby proposed and investigated the use of an internal osteosynthesis for stabilizing large bone defects. To this aim, a 3.5-mm-long segmental bone defect was induced in the mid-shaft of the femur using a Gigli saw and a jig. Bone fixation was performed using a titanium microlocking plate with four locking screws. The bone defect was either left empty or filled with a syngenic bone graft or filled with a coralline scaffold. Healing was monitored using radiographs. The healing process was further assessed using microcomputed tomography and histology 10 weeks after surgery. With the exception of one mouse that died during the surgical procedure, no complications were observed. A stable and reproducible bone fixation as well as a reproducible fixation of the implanted materials with full weight bearing was obtained in all animals tested. Nonunion was consistently observed in the group in which the defects were left empty. Bone union was obtained with the syngenic bone grafts, providing evidence that, although such defects were of critical size, bone healing was possible when the gold-standard material was used to fill the defect. Although new bone formation was greater in the coralline scaffold group than in the left-empty animal group, it remained limited and localized close to the bony edges, a consequence of the critical size of such bone defect. Our study established a reproducible, clinically relevant, femoral, atrophic nonunion, critical-sized defect, low morbidity mouse model. The present study was successful in designing and testing in a small animal model, a novel surgical method for the assessment of bone repair; this model has the potential to facilitate investigations of the molecular and cellular events involved in bone regeneration in load-bearing, segmental-bone defects.
Collapse
Affiliation(s)
- Mathieu Manassero
- Laboratory of Bioengineering and Biomechanics for Bone Articulation (B2OA–UMR CNRS 7052), University Paris-Diderot, Paris, France
| | | | | | | | | | | | | |
Collapse
|
21
|
Vaquette C, Fan W, Xiao Y, Hamlet S, Hutmacher DW, Ivanovski S. A biphasic scaffold design combined with cell sheet technology for simultaneous regeneration of alveolar bone/periodontal ligament complex. Biomaterials 2012; 33:5560-73. [DOI: 10.1016/j.biomaterials.2012.04.038] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 04/13/2012] [Indexed: 10/28/2022]
|
22
|
Zwingenberger S, Niederlohmann E, Vater C, Rammelt S, Matthys R, Bernhardt R, Valladares RD, Goodman SB, Stiehler M. Establishment of a femoral critical-size bone defect model in immunodeficient mice. J Surg Res 2012; 181:e7-e14. [PMID: 22765996 DOI: 10.1016/j.jss.2012.06.039] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 05/08/2012] [Accepted: 06/15/2012] [Indexed: 12/31/2022]
Abstract
BACKGROUND The development of innovative therapies for bone regeneration requires the use of advanced site-specific bone defect small-animal models. The achievement of proper fixation with a murine model is challenging due to the small dimensions of the murine femur. The aim of this investigation was to find the optimal defect size for a murine critical-size bone defect model using external fixation method. METHODS An external fixation device was attached to the right femur of 30 mice. Femoral bone defects of 1 mm (n = 10), 2 mm (n = 10), and 3 mm (n = 10) were created. Wounds were closed without any additional treatment. To investigate bone healing during the 12-wk observation period, x-ray analysis, histomorphology, immunohistochemistry, and μCT scans were performed. RESULTS MicroCT analyses after 12 wk showed that 3/8 1-mm defects, 5/8 2-mm defects, and 8/8 3-mm defects remained as nonunions. The defect volumes were 0.36 ± 0.42 mm³ (1-mm group), 1.40 ± 0.88 mm³ (2-mm group), and 2.88 ± 0.28 mm³ (3-mm group; P < 0.001, between all groups). CONCLUSION Using external fixation, a defect size of 3 mm is necessary to reliably create a persisting femoral bone defect in nude mice.
Collapse
Affiliation(s)
- Stefan Zwingenberger
- Department of Orthopedic Surgery, Stanford University, Stanford, California; Department of Orthopaedics and Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus at Technical University Dresden, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Srouji S, Ben-David D, Fromigué O, Vaudin P, Kuhn G, Müller R, Livne E, Marie PJ. Lentiviral-Mediated Integrin α5 Expression in Human Adult Mesenchymal Stromal Cells Promotes Bone Repair in Mouse Cranial and Long-Bone Defects. Hum Gene Ther 2012; 23:167-72. [DOI: 10.1089/hum.2011.059] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
- Samer Srouji
- Oral and Maxillofacial Surgery Department, Carmel Medical Center, 32000 Haifa, Israel
- Department of Anatomy and Cell Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Dror Ben-David
- Department of Anatomy and Cell Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Olivia Fromigué
- Laboratory of Osteoblast Biology and Pathology, Inserm U606, Paris, F-75475 France
- UMR 606, University Paris Diderot, Paris, F-75475 France
| | - Pascal Vaudin
- Inserm U966, Paris, F-75475 France
- University of Tours, Tours, F-37032 France
| | - Gisela Kuhn
- Institute for Biomechanics, ETH Zurich, 8093 Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, 8093 Zurich, Switzerland
| | - Erella Livne
- Department of Anatomy and Cell Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Pierre J. Marie
- Laboratory of Osteoblast Biology and Pathology, Inserm U606, Paris, F-75475 France
- UMR 606, University Paris Diderot, Paris, F-75475 France
| |
Collapse
|