1
|
Pu S, Fu R, Bertrand D, Willie BM, Yang H. A 4D time-lapse morphometry method to quantify bone formation and resorption during distraction osteogenesis. J Orthop Res 2025; 43:586-594. [PMID: 39511955 DOI: 10.1002/jor.26008] [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: 07/27/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024]
Abstract
Distraction osteogenesis (DO) is widely utilized for treating limb length discrepancy, nonunion, bone deformities and defects. This study sought to develop a 4D time-lapse morphometry method to quantify bone formation and resorption in mouse femur during DO based on image registration of longitudinal in vivo micro-CT scans. Female C57BL/6 mice (n = 7) underwent osteotomy, followed by 5 days of latency, 10 days of distraction and 35 days of consolidation. The mice were scanned with micro-CT at Days 5, 15, 25, 35, 45, and 50. Histological sectioning and Movat Pentachrome straining were performed at Day 50. After registration of two consecutive micro-CT images of the same bone (day x and day y), the spatially- and temporally-linked sequences of formation, resorption and quiescent bones at the distraction gap were identified and bone formation and resorption rates (BFRdayx-y and BRRdayx-y) were calculated. The overall percentage error of the registration method was 2.98% ± 0.89% and there was a strong correlation between histologically-measured bone area fraction and micro-CT-determined bone volume fraction at Day 50 (r = 0.89, p < 0.05). The 4D time-lapse morphometry indicated a rapid bone formation during the first 10 days of the consolidation phase (BFRday15-25 = 0.14 ± 0.05 mm3/day), followed by callus reshaping via equivalent bone formation and resorption rates. The 4D time-lapse morphometry method developed in this study allows for a continuous quantitative monitoring of the dynamic process of bone formation and resorption following distraction, which may offer a better understanding of the mechanism for mechano-regulated bone regeneration and aid for development of new treatment strategies of DO.
Collapse
Affiliation(s)
- Sishun Pu
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Ruisen Fu
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - David Bertrand
- Faculty of Dental Medicine and Oral Health Science, McGill University, Montreal, Canada
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada
| | - Bettina M Willie
- Faculty of Dental Medicine and Oral Health Science, McGill University, Montreal, Canada
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada
| | - Haisheng Yang
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| |
Collapse
|
2
|
Jaber M, Schmidt J, Kalkhof S, Gerstenfeld L, Duda GN, Checa S. OMIBONE: Omics-driven computer model of bone regeneration for personalized treatment. Bone 2024; 190:117288. [PMID: 39426580 DOI: 10.1016/j.bone.2024.117288] [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: 05/13/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
Treatment of bone fractures are standardized according to the AO classification, which mainly refers to the mechanical stabilization required in a given situation but neglect individual differences due to patient's healing potential or accompanying diseases. Specially in elderly or immune-compromised patients, the complexity of individual constrains on a biological as well as mechanical level are hard to account for. Here, we introduce a novel framework that allows to predict bone regeneration outcome using combined proteomic and mechanical analyses in a computer model. The framework uses Ingenuity Pathway Analysis (IPA) software to link protein changes to alterations in biological processes and integrates these in an Agent-Based Model (ABM) of bone regeneration. This combined framework allows to predict bone formation and the potential of an individual to heal a given fracture setting. The performance of the framework was evaluated by replicating the experimental setup of a mouse femur fracture stabilized with an intramedullary pin. The model was informed by serum derived proteomics data. The tissue formation patterns were compared against experimental data based on x-ray and histology images. The results indicate the framework potential in predicting an individual's bone formation potential and hold promise as a concept to enable personalized bone healing predictions for a chosen fracture fixation.
Collapse
Affiliation(s)
- Mahdi Jaber
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - Johannes Schmidt
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Stefan Kalkhof
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Louis Gerstenfeld
- Department of Orthopaedic Surgery, Boston University of Medicine, Boston, MA, United States of America
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany.
| |
Collapse
|
3
|
Kim H, Kang KW, Chekalkin T, Park JW, Chung HK, Kang BJ, Choi SW. Pilot study on the feasibility of shape memory alloy implantation for Vancouver type B1 periprosthetic femoral fractures in a canine model: a step toward advancing treatment modalities. J Orthop Surg Res 2024; 19:510. [PMID: 39192290 DOI: 10.1186/s13018-024-05011-4] [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: 02/22/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Cerclage wiring is commonly used for treating fractures; however, it has several limitations, including mechanical weakness, decreased blood circulation, and technical complexity. In this study, we developed an implant using a shape memory alloy (SMA) and tested its efficacy in treating Vancouver type B1 (VB1) periprosthetic femoral fractures (PFFs) in a canine model. METHODS The mid-diaphyseal fracture models underwent reduction via the SMA plate (SMA group) or the cerclage cable plate (cable group) method in randomly selected pelvic limbs. An intraoperative evaluation was conducted to assess the surgical time and difficulty related to implant fitting. Clinical assessments, radiography, microcomputed tomography (micro-CT), histological analysis, positron emission tomography (PET)/CT, and galvanic corrosion analysis were conducted for 52 weeks to evaluate bone healing and blood perfusion. RESULTS The results for bone healing and blood perfusion were not significantly different between the groups (p > 0.05). In addition, no evidence of galvanic corrosion was present in any of the implants. However, the median surgical time was 75 min (range, 53-82 min) for the SMA group and 126 min (range, 120-171 min) for the cable group, which was a statistically significant difference (p = 0.0286). CONCLUSIONS This study assessed the ability of a newly developed shape memory alloy (SMA) to treat VB1 periprosthetic femoral fractures (PFFs) in canines for over a 52-week period and revealed outcomes comparable to those of traditional methods in terms of bone healing and mechanical stability. Despite the lower surgical complexity and potential time-saving benefits of this treatment, further research is needed to confirm its efficacy.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
- BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, Korea
| | - Kyu-Won Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
| | - Timofey Chekalkin
- Research and Development Center, TiNiKo Company, Limited, Osong, 28164, Korea
| | - Jang-Woo Park
- Korea Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Hye-Kyung Chung
- Korea Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea.
- BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, Korea.
| | - Sung-Woo Choi
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea.
- Department of Orthopedic Surgery, Soonchunhyang University College of Medicine, Seoul, 04401, Korea.
| |
Collapse
|
4
|
Ren T, Inglis B, Darwiche S, Dailey HL. Torsion constants and virtual mechanical tests are valid image-based surrogate measures of ovine fracture healing. J Orthop Res 2024; 42:1810-1819. [PMID: 38491964 DOI: 10.1002/jor.25836] [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: 09/30/2022] [Revised: 08/30/2023] [Accepted: 02/26/2024] [Indexed: 03/18/2024]
Abstract
In large animal studies, the mechanical reintegration of the bone fragments is measured using postmortem physical testing, but these assessments can only be performed once, after sacrifice. Image-based virtual mechanical testing is an attractive alternative because it could be used to monitor healing longitudinally. However, the procedures and software required to perform finite element analysis (FEA) on subject-specific models for virtual mechanical testing can be time consuming and costly. Accordingly, the goal of this study was to determine whether a simpler image-based geometric measure-the torsion constant, sometimes known as polar moment of inertia-can be reliably used as a surrogate measure of bone healing in large animals. To achieve this, postmortem biomechanical testing and microCT scans were analyzed for a total of 33 operated and 20 intact ovine tibiae. An image-processing procedure to compute the attenuation-weighted torsion constant from the microCT scans was developed in MATLAB and this code has been made freely available. Linear regression analysis was performed between the postmortem biomechanical data, the results of virtual mechanical testing using FEA, and the torsion constants measured from the scans. The results showed that virtual mechanical testing is the most reliable surrogate measure of postmortem torsional rigidity, having strong correlations and high absolute agreement. However, when FEA is not practical, the torsion constant is a viable alternative surrogate measure that is moderately correlated with postmortem torsional rigidity and can be readily calculated.
Collapse
Affiliation(s)
- Tianyi Ren
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Brendan Inglis
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Salim Darwiche
- Musculoskeltal Research Unit (MSRU), Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
| | - Hannah L Dailey
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania, USA
| |
Collapse
|
5
|
Inoue S, Li C, Hatakeyama J, Jiang H, Kuroki H, Moriyama H. Higher-intensity ultrasound accelerates fracture healing via mechanosensitive ion channel Piezo1. Bone 2023; 177:116916. [PMID: 37777037 DOI: 10.1016/j.bone.2023.116916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
Osteoporosis-related fractures are a major public health problem. Mechanobiological stimulation utilizing low-intensity pulsed ultrasound (LIPUS) is the most widely accepted modality for accelerating fracture healing. However, recent evidence has demonstrated the ineffectiveness of LIPUS, and the biophysical mechanisms of ultrasound-induced bone formation also remain elusive. Here, we demonstrate that ultrasound at a higher intensity than LIPUS effectively accelerates fracture healing in a mouse osteoporotic fracture model. Higher-intensity ultrasound promoted chondrogenesis and hypertrophic differentiation of chondrocytes in the fracture callus. Higher-intensity ultrasound also increased osteoblasts and newly formed bone in the callus, resulting in accelerated endochondral ossification during fracture healing. In addition, we found that accelerated fracture healing by ultrasound exposure was attenuated when the mechanosensitive ion channel Piezo1 was inhibited by GsMTx4. Ultrasound-induced new bone formation in the callus was attenuated in fractured mice treated with GsMTx4. Similar results were also confirmed in a 3D osteocyte-osteoblast co-culture system, where osteocytic Piezo1 knockdown attenuated the expression of osteoblastic genes after ultrasound exposure. Together these results demonstrate that higher-intensity ultrasound than clinically used LIPUS can accelerate endochondral ossification after fractures. Furthermore, our results suggest that mechanotransduction via Piezo1 mediates ultrasound-stimulated fracture healing and bone formation.
Collapse
Affiliation(s)
- Shota Inoue
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Changxin Li
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Junpei Hatakeyama
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan; Research Fellowship of the Japan Society for the Promotion of Science, Japan
| | - Hanlin Jiang
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Hiroshi Kuroki
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideki Moriyama
- Life and Medical Sciences Area, Health Sciences Discipline, Kobe University, Kobe, Japan.
| |
Collapse
|
6
|
Shih YV, Kingsley D, Newman H, Hoque J, Gupta A, Lascelles BDX, Varghese S. Multi-Functional Small Molecule Alleviates Fracture Pain and Promotes Bone Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303567. [PMID: 37939302 PMCID: PMC10754086 DOI: 10.1002/advs.202303567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/21/2023] [Indexed: 11/10/2023]
Abstract
Bone injuries such as fractures are one major cause of morbidities worldwide. A considerable number of fractures suffer from delayed healing, and the unresolved acute pain may transition to chronic and maladaptive pain. Current management of pain involves treatment with NSAIDs and opioids with substantial adverse effects. Herein, we tested the hypothesis that the purine molecule, adenosine, can simultaneously alleviate pain and promote healing in a mouse model of tibial fracture by targeting distinctive adenosine receptor subtypes in different cell populations. To achieve this, a biomaterial-assisted delivery of adenosine is utilized to localize and prolong its therapeutic effect at the injury site. The results demonstrate that local delivery of adenosine inhibited the nociceptive activity of peripheral neurons through activation of adenosine A1 receptor (ADORA1) and mitigated pain as demonstrated by weight bearing and open field movement tests. Concurrently, local delivery of adenosine at the fracture site promoted osteogenic differentiation of mesenchymal stromal cells through adenosine A2B receptor (ADORA2B) resulting in improved bone healing as shown by histological analyses and microCT imaging. This study demonstrates the dual role of adenosine and its material-assisted local delivery as a feasible therapeutic approach to treat bone trauma and associated pain.
Collapse
Affiliation(s)
- Yu‐Ru V. Shih
- Department of Orthopaedic SurgeryDuke University School of MedicineDurhamNC27710USA
| | - David Kingsley
- Department of Orthopaedic SurgeryDuke University School of MedicineDurhamNC27710USA
| | - Hunter Newman
- Department of Mechanical Engineering and Materials ScienceDuke UniversityDurhamNC27710USA
| | - Jiaul Hoque
- Department of Orthopaedic SurgeryDuke University School of MedicineDurhamNC27710USA
| | - Ankita Gupta
- Translational Research in Pain ProgramDepartment of Clinical SciencesCollege of Veterinary MedicineNorth Carolina State UniversityRaleighNC27607USA
| | - B. Duncan X. Lascelles
- Translational Research in Pain ProgramDepartment of Clinical SciencesCollege of Veterinary MedicineNorth Carolina State UniversityRaleighNC27607USA
- Thurston Arthritis CenterUniversity of North Carolina School of MedicineChapel HillNC27599USA
- Center for Translational Pain MedicineDepartment of AnesthesiologyDuke University School of MedicineDurhamNC27710USA
- Comparative Pain Research and Education CenterCollege of Veterinary MedicineNorth Carolina State UniversityRaleighNC27607USA
| | - Shyni Varghese
- Department of Orthopaedic SurgeryDuke University School of MedicineDurhamNC27710USA
- Department of Mechanical Engineering and Materials ScienceDuke UniversityDurhamNC27710USA
- Department of Biomedical EngineeringDuke UniversityDurhamNC27710USA
| |
Collapse
|
7
|
Jeong CH, Lim SY, Um JE, Lim HW, Hwang KH, Park KM, Yun JS, Kim D, Huh JK, Kim HS, Yook JI, Kim NH, Kwak YH. Micellized protein transduction domain-bone morphogenetic protein-2 accelerates bone healing in a rat tibial distraction osteogenesis model. Acta Biomater 2023; 170:360-375. [PMID: 37611691 DOI: 10.1016/j.actbio.2023.08.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023]
Abstract
The clinical application of growth factors such as recombinant human bone morphogenetic protein-2 (rh-BMP-2), for functional bone regeneration remains challenging due to limited in vivo efficacy and adverse effects of previous modalities. To overcome the instability and short half-life of rh-BMP-2 in vivo, we developed a novel osteogenic supplement by fusing a protein transduction domain (PTD) with BMP-2, effectively creating a prodrug of BMP-2. In this study, we first created an improved PTD-BMP-2 formulation using lipid nanoparticle (LNP) micellization, resulting in downsizing from micrometer to nanometer scale and achieving a more even distribution. The micellized PTD-BMP-2 (mPTD-BMP-2) demonstrated improved distribution and aggregation profiles. As a prodrug of BMP-2, mPTD-BMP-2 successfully activated Smad1/5/8 and induced mineralization with osteogenic gene induction in vitro. In vivo pharmacokinetic analysis revealed that mPTD-BMP-2 had a much more stable pharmacokinetic profile than rh-BMP-2, with a 7.5-fold longer half-life. The in vivo BMP-responsive element (BRE) reporter system was also successfully activated by mPTD-BMP-2. In the in vivo rat tibia distraction osteogenesis (DO) model, micro-computed tomography (micro-CT) scan findings indicated that mPTD-BMP-2 significantly increased bone volume, bone surface, axis moment of inertia (MOI), and polar MOI. Furthermore, it increased the expression of osteogenesis-related genes, and induced bone maturation histologically. Based on these findings, mPTD-BMP-2 could be a promising candidate for the next-generation osteogenesis drug to promote new bone formation in DO surgery. STATEMENT OF SIGNIFICANCE: This study introduces micellized bone morphogenetic protein-2 (mPTD-BMP-2), a next-generation osteogenic supplement that combines protein transduction domain (PTD) and nano-sized micelle formulation technique to improve transduction efficiency and stability. The use of PTD represents a novel approach, and our results demonstrate the superiority of mPTD-BMP-2 over rh-BMP-2 in terms of in vivo pharmacokinetic profile and osteogenic potential, particularly in a rat tibial model of distraction osteogenesis. These findings have significant scientific impact and potential clinical applications in the treatment of bone defects that require distraction osteogenesis. By advancing the field of osteogenic supplements, our study has the potential to contribute to the development of more effective treatments for musculoskeletal disorders.
Collapse
Affiliation(s)
- Cheol Hee Jeong
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, 03722, Korea; Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Song-Yi Lim
- Department of Orthopedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul, 05505, Korea
| | - Jo Eun Um
- MET Life Science, Seoul, 03722, Korea
| | - Hyo Won Lim
- Department of Orthopedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul, 05505, Korea
| | | | - Kyeong-Mee Park
- Department of Advanced General Dentistry, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Jun Seop Yun
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, 03722, Korea; Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Dohun Kim
- Department of Orthopedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul, 05505, Korea
| | - Jong-Ki Huh
- Department of Oral and Maxillofacial Surgery, Gangnam Severance Hospital, Yonsei University College of Dentistry, Seoul, 06273, Korea
| | - Hyun Sil Kim
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, 03722, Korea; Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea; MET Life Science, Seoul, 03722, Korea
| | - Jong In Yook
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, 03722, Korea; Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea; MET Life Science, Seoul, 03722, Korea
| | - Nam Hee Kim
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea; MET Life Science, Seoul, 03722, Korea.
| | - Yoon Hae Kwak
- Department of Orthopedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul, 05505, Korea.
| |
Collapse
|
8
|
Hopkinson M, Jones G, Evans L, Gohin S, Magnusdottir R, Salmon P, Chenu C, Meeson R, Javaheri B, Pitsillides AA. A new method for segmentation and analysis of bone callus in rodent fracture models using micro-CT. J Orthop Res 2023; 41:1717-1728. [PMID: 36582023 PMCID: PMC10947128 DOI: 10.1002/jor.25507] [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: 01/26/2022] [Revised: 10/18/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022]
Abstract
Fracture burden has created a need to better understand bone repair processes under different pathophysiological states. Evaluation of structural and material properties of the mineralized callus, which is integral to restoring biomechanical stability is, therefore, vital. Microcomputed tomography (micro-CT) can facilitate noninvasive imaging of fracture repair, however, current methods for callus segmentation are only semiautomated, restricted to defined regions, time/labor intensive, and prone to user variation. Herein, we share a new automatic method for segmenting callus in micro-CT tomograms that will allow for objective, quantitative analysis of the bone fracture microarchitecture. Fractured and nonfractured mouse femurs were scanned and processed by both manual and automated segmentation of fracture callus from cortical bone after which microarchitectural parameters were analyzed. All segmentation and analysis steps were performed using CTAn (Bruker) with automatic segmentation performed using the software's image-processing plugins. Results showed automatic segmentation reliably and consistently segmented callus from cortical bone, demonstrating good agreement with manual methods with low bias: tissue volume (TV): -0.320 mm3 , bone volume (BV): 0.0358 mm3 , and bone volume/tissue volume (BV/TV): -3.52%, and was faster and eliminated user-bias and variation. Method scalability and translatability across rodent models were verified in scans of fractured rat femora showing good agreement with manual methods with low bias: TV: -3.654 mm3 , BV: 0.830 mm3 , and BV/TV: 7.81%. Together, these data validate a new automated method for segmentation of callus and cortical bone in micro-CT tomograms that we share as a fast, reliable, and less user-dependent tool for application to study bone callus in fracture, and potentially elsewhere.
Collapse
Affiliation(s)
- Mark Hopkinson
- Comparative Biological SciencesRoyal Veterinary CollegeLondonUK
| | - Gareth Jones
- Clinical Science and ServicesRoyal Veterinary CollegeLondonUK
| | - Lucinda Evans
- Comparative Biological SciencesRoyal Veterinary CollegeLondonUK
| | - Stephanie Gohin
- Comparative Biological SciencesRoyal Veterinary CollegeLondonUK
| | | | | | - Chantal Chenu
- Comparative Biological SciencesRoyal Veterinary CollegeLondonUK
| | - Richard Meeson
- Clinical Science and ServicesRoyal Veterinary CollegeLondonUK
- Division of SurgeryUniversity College LondonLondonUK
| | - Behzad Javaheri
- Comparative Biological SciencesRoyal Veterinary CollegeLondonUK
- Present address:
School of Mathematics, Computer Science and EngineeringCity University of LondonLondonUK
| | | |
Collapse
|
9
|
Nurrachman AS, Azhari A, Epsilawati L, Pramanik F. Temporal Pattern of micro-CT Angiography Vascular Parameters and VEGF mRNA Expression in Fracture Healing: a Radiograph and Molecular Comparison. Eur J Dent 2023. [PMID: 36716788 DOI: 10.1055/s-0042-1757466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis plays an important role in fracture healing with vascular endothelial growth factor (VEGF) as the main protein involved. Micro-computed tomography (CT) angiography may be used to analyze this revascularization with several parameters such as number of branches, total volume, and diameter. This systematic review is aimed to assess available studies on the temporal pattern of vascular imaging on micro-CT angiographs, especially in terms of the number of branches, total volume, and diameter as well as the temporal pattern of VEGF mRNA expression as the molecular comparison during bone fracture healing. This review was conducted according to the PRISMA guidelines. Electronic database searches were performed using PubMed, ProQuest, ScienceDirect, EBSCOhost, Taylor & Francis Online, and hand searching. The search strategy and keywords were adjusted to each database using the Boolean operators and other available limit functions to identify most relevant articles based on our inclusion and exclusion criteria. Screening and filtration were done in several stages by removing the duplicates and analyzing each title, abstract, and full-text in all included entries. Data extraction was done for syntheses to summarize the temporal pattern of each parameter. A total of 28 articles were eligible and met all criteria, 11 articles were synthesized in its angiograph's analysis, 16 articles were synthesized in its VEGF mRNA expression analysis, and 1 article had both parameters analyzed. The overall temporal pattern of both three micro-CT angiographic parameters and VEGF mRNA expression was in line qualitatively. The number of branches, total volume, and diameter of the blood vessels in micro-CT angiography showed an exponential rise at week 2 and decline at week 3 of fracture healing, with the VEGF mRNA expression concurrently showing a consistent pattern in the phase.
Collapse
Affiliation(s)
- Aga Satria Nurrachman
- Department of Oral and Maxillofacial Radiology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Azhari Azhari
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, West Java, Indonesia
| | - Lusi Epsilawati
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, West Java, Indonesia
| | - Farina Pramanik
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, West Java, Indonesia
| |
Collapse
|
10
|
Bottagisio M, Palombella S, Lopa S, Sangalli F, Savadori P, Biagiotti M, Sideratou Z, Tsiourvas D, Lovati AB. Vancomycin-nanofunctionalized peptide-enriched silk fibroin to prevent methicillin-resistant Staphylococcus epidermidis-induced femoral nonunions in rats. Front Cell Infect Microbiol 2023; 12:1056912. [PMID: 36683682 PMCID: PMC9851397 DOI: 10.3389/fcimb.2022.1056912] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Implant-related infections and infected fractures are significant burdens in orthopedics. Staphylococcus epidermidis is one of the main causes of bone infections related to biofilm formation upon implants. Current antibiotic prophylaxis/therapy is often inadequate to prevent biofilm formation and results in antibiotic resistance. The development of bioactive materials combining antimicrobial and osteoconductive properties offers great potential for the eradication of microorganisms and for the enhancement of bone deposition in the presence of infections. The purpose of this study is to prevent the development of methicillin-resistant S. epidermidis (MRSE)-infected nonunion in a rat model. Methods To this end, a recently developed in our laboratories bioactive material consisting of antibiotic-loaded nanoparticles based on carboxylic acid functionalized hyperbranched aliphatic polyester (CHAP) that are integrated into peptide-enriched silk fibroin sponges with osteoconductive properties (AFN-PSF) was employed, whose biocompatibility and microbiological tests provided proof of its potential for the treatment of both orthopedic and dental infections. In particular, non-critical femoral fractures fixed with plates and screws were performed in Wistar rats, which were then randomly divided into three groups: 1) the sham control (no infection, no treatment); 2) the control group, infected with MRSE and treated with peptide-enriched silk fibroin sponges incorporating non-drug-loaded functionalized nanoparticles (PSF); 3) the treated group, infected with MRSE and treated with peptide-enriched silk fibroin sponges incorporating vancomycin-loaded functionalized nanoparticles (AFN-PSF). After 8 weeks, bone healing and osteomyelitis were clinically assessed and evaluated by micro-CT, microbiological and histological analyses. Results The sham group showed no signs of infection and complete bone healing. The PSF group failed to repair the infected fracture, displaying 75% of altered bone healing and severe signs of osteomyelitis. The AFN-PSF treated group reached 70% of fracture healing in the absence of signs of osteomyelitis, such as abscesses in the cortical and intraosseous compartments and bone necrosis with sequestra. Discussion AFN-PSF sponges have proven effective in preventing the development of infected nonunion in vivo. The proposed nanotechnology for local administration of antibiotics can have a significant impact on patient health in the case of orthopedic infections.
Collapse
Affiliation(s)
- Marta Bottagisio
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Clinical Chemistry and Microbiology, Milan, Italy
| | - Silvia Palombella
- IRCCS Istituto Ortopedico Galeazzi, Cell and Tissue Engineering Laboratory, Milan, Italy
| | - Silvia Lopa
- IRCCS Istituto Ortopedico Galeazzi, Cell and Tissue Engineering Laboratory, Milan, Italy
| | - Fabio Sangalli
- IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Laboratory of Renal Biophysics, Department of Biomedical Engineering, Bergamo, Italy
| | - Paolo Savadori
- IRCCS Istituto Ortopedico Galeazzi, Department of Endodontics, Milan, Italy
| | | | - Zili Sideratou
- National Centre for Scientific Research "Demokritos", Institute of Nanoscience and Nanotechnology, Aghia Paraskevi, Greece
| | - Dimitris Tsiourvas
- National Centre for Scientific Research "Demokritos", Institute of Nanoscience and Nanotechnology, Aghia Paraskevi, Greece
| | - Arianna B Lovati
- IRCCS Istituto Ortopedico Galeazzi, Cell and Tissue Engineering Laboratory, Milan, Italy
| |
Collapse
|
11
|
Viero A, Biehler-Gomez L, Messina C, Cappella A, Giannoukos K, Viel G, Tagliaro F, Cattaneo C. Utility of micro-CT for dating post-cranial fractures of known post-traumatic ages through 3D measurements of the trabecular inner morphology. Sci Rep 2022; 12:10543. [PMID: 35732857 DOI: 10.1038/s41598-022-14530-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/08/2022] [Indexed: 11/09/2022] Open
Abstract
Fracture dating is an issue at the forefront of forensic sciences. While dating fracture is crucial to understanding and verifying the chronology of events in cases of abuse and violent death, its application is the subject of considerable discussion in the scientific community, filled with limitations and difficulties. Current methods for fracture dating are mainly based on a qualitative assessment through macroscopy, microscopy, and imaging and subject to variations depending on the experience of the observer. In this paper, we investigated the potential of quantifiable micro-CT analysis for fracture dating. Five histomorphometric parameters commonly used for the study of the 3D bone trabecular microarchitecture with micro-CT were calculated based on nine fractures of known post-traumatic ages, including the degree of anisotropy, connectivity density, bone volume fraction, trabecular thickness, and trabecular separation. As a result, trends in the evolution of the microarchitecture of the bone relative to age of the callus could be identified, in particular concerning anisotropy, trabecular separation and connectivity density, consistent with the healing bone process. The findings obtained in this pilot study encourage further research in quantifiable parameters of the bone microarchitecture as they could represent useful features for the construction of objective models for fracture dating.
Collapse
Affiliation(s)
- Alessia Viero
- Legal Medicine and Toxicology Unit, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy.,Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, P.le Scuro, 10, 37134, Verona, Italy.,Laboratorio Di Antropologia E Odontologia Forense (LABANOF), Sezione Di Medicina Legale, Department of Biomedical Sciences for Health, Università Degli Studi Di Milano, Via Mangiagalli 37, 20133, Milan, Italy
| | - Lucie Biehler-Gomez
- Laboratorio Di Antropologia E Odontologia Forense (LABANOF), Sezione Di Medicina Legale, Department of Biomedical Sciences for Health, Università Degli Studi Di Milano, Via Mangiagalli 37, 20133, Milan, Italy.
| | - Carmelo Messina
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Annalisa Cappella
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,Laboratorio Di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Konstantinos Giannoukos
- Elettra-Sincrotrone Trieste S.C.P.A., SYRMEP Group, AREA Science Park, Strada Statale 14, 34149, Basovizza, Trieste, Italy
| | - Guido Viel
- Legal Medicine and Toxicology Unit, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Franco Tagliaro
- Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, P.le Scuro, 10, 37134, Verona, Italy.,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Cristina Cattaneo
- Laboratorio Di Antropologia E Odontologia Forense (LABANOF), Sezione Di Medicina Legale, Department of Biomedical Sciences for Health, Università Degli Studi Di Milano, Via Mangiagalli 37, 20133, Milan, Italy
| |
Collapse
|
12
|
Zhu D, Fang H, Yu H, Liu P, Yang Q, Luo P, Zhang C, Gao Y, Chen YX. Alcohol-induced inhibition of bone formation and neovascularization contributes to the failure of fracture healing via the miR-19a-3p/FOXF2 axis. Bone Joint Res 2022; 11:386-397. [PMID: 35730670 PMCID: PMC9233406 DOI: 10.1302/2046-3758.116.bjr-2021-0596.r1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Aims Alcoholism is a well-known detrimental factor in fracture healing. However, the underlying mechanism of alcohol-inhibited fracture healing remains poorly understood. Methods MicroRNA (miR) sequencing was performed on bone mesenchymal stem cells (BMSCs). The effects of alcohol and miR-19a-3p on vascularization and osteogenic differentiation were analyzed in vitro using BMSCs and human umbilical vein endothelial cells (HUVECs). An in vivo alcohol-fed mouse model of femur fracture healing was also established, and radiological and histomorphometric analyses were used to evaluate the role of miR-19a-3p. The binding of miR-19a-3p to forkhead box F2 (FOXF2) was analyzed using a luciferase reporter assay. Results miR-19a-3p was identified as one of the key regulators in the osteogenic differentiation of BMSCs, and was found to be downregulated in the alcohol-fed mouse model of fracture healing. In vitro, miR-19a-3p expression was downregulated after ethanol administration in both BMSCs and HUVECs. Vascularization and osteogenic differentiation were independently suppressed by ethanol and reversed by miR-19a-3p. In addition, the luciferase reporter assay showed that FOXF2 is the direct binding target of miR-19a-3p. In vivo, miR-19a-3p agomir stimulated callus transformation and improved the alcohol-impaired fracture healing. Conclusion This study is the first to demonstrate that the miR-19a-3p/FOXF2 axis has a pivotal role in alcohol-impaired fracture healing, and may be a potential therapeutic target. Cite this article: Bone Joint Res 2022;11(6):386–397.
Collapse
Affiliation(s)
- Daoyu Zhu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Haoyu Fang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hongping Yu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Pei Liu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qianhao Yang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Pengbo Luo
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yi-Xuan Chen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
13
|
Correa KDP, Silva MET, Ribeiro OS, Matta SLP, Peluzio MDCG, Oliveira EB, Coimbra JSDR. Homogenised and pasteurised human milk: lipid profile and effect as a supplement in the enteral diet of Wistar rats. Br J Nutr 2022; 127:711-721. [PMID: 33902762 DOI: 10.1017/s0007114521001380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The retention of human milk (HM) fat in nasogastric probes of infusion pumps can be observed during the feed of infants unable to suck at the mother's breast. The lack of homogenisation of HM could contribute to the fat holding. Therefore, the present study evaluated (i) the influence of homogenisation on milk fat retaining in infant feeding probes and (ii) the in vivo effect of the homogenisation on lipid absorption by Wistar rats. The animals were fed with HM treated following two processing conditions, that is, pasteurised and homogenised-pasteurised. The animals were randomly subdivided into four experimental groups: water-fed (control), pasteurised milk, homogenised-pasteurised milk and pasteurised-skimmed milk. The results of food consumption, mass body gain, corporate metrics and plasma blood levels of total cholesterol did not show any difference (P < 0·05) among the three types of HM used in the experiments. The liver, intestine and intra-abdominal adipose tissue of the four groups of animals presented normal and healthy histology. The composition of fatty acids in the brain tissue of animals fed with homogenised HM increased when compared with the groups fed with non-homogenised HM. These values were 11·08 % higher for arachidonic acids, 6·59 % for DAH and 47·92 % for nervous acids. The ingestion of homogenised HM promoted higher absorption of milk nutrients. Therefore, the addition of the homogenisation stage in HM processing could be an alternative to reduce fat retention in probes and to improve the lipids' absorption in the body.
Collapse
Affiliation(s)
- Kely de Paula Correa
- Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa (UFV), Av. P.H. Rolfs, s/n, 36570-900 Viçosa, MG, Brasil
| | - Monique E T Silva
- Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa (UFV), Av. P.H. Rolfs, s/n, 36570-900 Viçosa, MG, Brasil
| | - Otávio S Ribeiro
- Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa (UFV), Av. P.H. Rolfs, s/n, 36570-900 Viçosa, MG, Brasil
| | - Sérgio L P Matta
- Departamento de Biologia Vegetal, Universidade Federal de Viçosa (UFV), Av. P.H. Rolfs, s/n, 36570-900 Viçosa, MG, Brasil
| | - Maria do Carmo G Peluzio
- Departamento de Nutrição e Saúde, Universidade Federal de Viçosa (UFV), Av. P.H. Rolfs, s/n, 36570-900 Viçosa, MG, Brasil
| | - Eduardo B Oliveira
- Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa (UFV), Av. P.H. Rolfs, s/n, 36570-900 Viçosa, MG, Brasil
| | - Jane S Dos R Coimbra
- Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa (UFV), Av. P.H. Rolfs, s/n, 36570-900 Viçosa, MG, Brasil
| |
Collapse
|
14
|
Knox AM, McGuire AC, Natoli RM, Kacena MA, Collier CD. Methodology, selection, and integration of fracture healing assessments in mice. J Orthop Res 2021; 39:2295-2309. [PMID: 34436797 PMCID: PMC8542592 DOI: 10.1002/jor.25172] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 02/04/2023]
Abstract
Long bone fractures are one of the most common and costly medical conditions encountered after trauma. Characterization of the biology of fracture healing and development of potential medical interventions generally involves animal models of fracture healing using varying genetic or treatment groups, then analyzing relative repair success via the synthesis of diverse assessment methodologies. Murine models are some of the most widely used given their low cost, wide variety of genetic variants, and rapid breeding and maturation. This review addresses key concerns regarding fracture repair investigations in mice and may serve as a guide in conducting and interpreting such studies. Specifically, this review details the procedures, highlights relevant parameters, and discusses special considerations for the selection and integration of the major modalities used for quantifying fracture repair in such studies, including X-ray, microcomputed tomography, histomorphometric, biomechanical, gene expression and biomarker analyses.
Collapse
Affiliation(s)
- Adam M. Knox
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Anthony C. McGuire
- 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
| | | |
Collapse
|
15
|
Viero A, Obertová Z, Cappella A, Messina C, Sconfienza LM, Sardanelli F, Tritella S, Montisci M, Gregori D, Tagliaro F, Cattaneo C. The problem of dating fractures: A retrospective observational study of radiologic features of fracture healing in adults. Forensic Sci Int 2021; 329:111058. [PMID: 34710653 DOI: 10.1016/j.forsciint.2021.111058] [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: 04/15/2021] [Revised: 09/11/2021] [Accepted: 10/14/2021] [Indexed: 11/18/2022]
Abstract
Accurate dating of bone fractures can be crucial in the context of forensic trauma analysis since it may provide essential information for the corroboration or contradiction of statements by victims or suspects in cases of domestic abuse or torture. The different stages of bone healing have been well described in the existing literature, and some previous studies attempted to define related timelines particularly in the paediatric population. However, the bone healing process can be very variable and despite the importance of the topic in the forensic field, so far little is known about the radiological appearance of bone fractures at different healing stages, and how this correlates with time, especially in an adult population. The aim of this retrospective observational study was to describe the temporal aspect of pre-defined healing stages as they appear on radiographs of tubular bone fractures in adults, and explore the effect of potential cofounding variables. A dynamic nomogram was developed as a user-friendly tool to be eventually applied in clinical or medico-legal settings. This study showed that the posttraumatic time interval (PTTI) increased progressively with the pre-defined healing stages. However, confounding factors, such as patients' age, sex, and location of the fracture need to be accounted for in the final estimation model. Further studies are needed to explore more potentially confounding variables to refine the presented outcomes. Better knowledge of the effect of different confounding variables in the dating of fracture healing will contribute to greater accuracy of PTTI estimation of bone fractures in adults.
Collapse
Affiliation(s)
- Alessia Viero
- Legal Medicine and Toxicology Unit, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy; Laboratorio di Antropologia e Odontologia Forense (LABANOF), Sezione di Medicina Legale, Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, P.le Scuro, 10-37134, Verona, Italy
| | - Zuzana Obertová
- Laboratorio di Antropologia e Odontologia Forense (LABANOF), Sezione di Medicina Legale, Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; Centre for Forensic Anthropology, School of Social Sciences, The University of Western Australia, Australia
| | - Annalisa Cappella
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, Italy; Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.
| | - Carmelo Messina
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Luca Maria Sconfienza
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Francesco Sardanelli
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; IRCCS Policlinico San Donato, Milan, Italy
| | - Stefania Tritella
- Unit of Radiology, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Massimo Montisci
- Legal Medicine and Toxicology Unit, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Dario Gregori
- Unit of Biostatistics, Epidemiology and Public Health - Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Università degli Studi di Padova, Padova, Veneto, Italy
| | - Franco Tagliaro
- Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, P.le Scuro, 10-37134, Verona, Italy; World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Cristina Cattaneo
- Laboratorio di Antropologia e Odontologia Forense (LABANOF), Sezione di Medicina Legale, Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
16
|
The effect of piezocision vs no piezocision on maxillary extraction space closure: A split-mouth, randomized controlled clinical trial. Am J Orthod Dentofacial Orthop 2021; 161:7-19.e2. [PMID: 34654603 DOI: 10.1016/j.ajodo.2021.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The aim of this 2-arm-parallel, split-mouth trial was to investigate the effects of piezocision compared with no piezocision on maxillary canine distalization and to evaluate patient perceptions on the surgical procedure. METHODS Twenty-two participants requiring extractions of maxillary first premolars were recruited from the Department of Orthodontics (Sydney Dental Hospital) waiting list. After leveling and alignment, a minimum of 3 mm space was required for canine retraction. Piezocision cuts distal to the canines were 4 mm long and 3 mm deep into the buccal cortical plate. The canine retraction was initiated on both sides immediately after surgery, with coil springs delivering 150 g of force per side. Random assignment of piezocision or control intervention on the patient's right side was performed (www.randomisation.com) for the random number generation, and allocation concealment was accomplished with opaque, sealed envelopes. Patients were assessed every 6 weeks for coil activation and alginate impressions over 18 weeks. The primary outcome was the amount of tooth movement in mm. Secondary outcomes were canine rotation, anchorage loss measured on scanned dental models, and patient pain levels and perception on piezocision using visual analog scale questionnaires. Blinding was feasible for the dental model measurements. RESULTS Twenty patients completed the trial. The treatment × time interaction showed no statistically or clinically significant differences in maxillary extraction space closure (b = -0.02; 95% confidence interval [CI], -0.29 to 0.25; P = 0.89) canine rotation (b = -1.45; 95% CI, -4 to 1.09; P = 0.26) and anchorage loss (b = -0.02; 95% CI, -0.38 to 0.34; P = 0.92). All patients except for one had minimal pain associated with the piezocision surgery but found the procedure tolerable and would recommend it. No harm occurred during the trial. CONCLUSIONS Piezocision-assisted maxillary canine distalization was similar to distalization with conventional orthodontics with patients tolerating the procedure.
Collapse
|
17
|
Vovos TJ, Furman BD, Huebner JL, Kimmerling KA, Utturkar GM, Green CL, Kraus VB, Guilak F, Olson SA. Initial displacement of the intra-articular surface after articular fracture correlates with PTA in C57BL/6 mice but not "superhealer" MRL/MpJ mice. J Orthop Res 2021; 39:1977-1987. [PMID: 33179316 DOI: 10.1002/jor.24912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/08/2020] [Accepted: 11/08/2020] [Indexed: 02/04/2023]
Abstract
Posttraumatic arthritis (PTA) occurs commonly after articular fracture and may arise, in part, from joint surface incongruity after injury. MRL/MpJ (MRL) "super-healer" mice are protected from PTA compared to C57BL/6 (B6) mice following articular fracture. However, the relationship between the initial displacement of the articular surface, biologic response, and susceptibility to PTA after fracture remains unclear. The objective of this study was to assess whether joint incongruity after articular fracture, as measured by in vivo micro-computed tomography (microCT), could predict pathomechanisms of PTA in mice. B6 and MRL mice (n = 12/strain) received a closed articular fracture (fx) of the left tibial plateau. Articular incongruity was quantified as bone surface deviations (BSD) for each in vivo microCT scan obtained from pre-fx to 8 weeks post-fx, followed by histologic assessment of arthritis. Serum concentrations of bone formation (PINP) and bone resorption (CTX-I) biomarkers were quantified longitudinally. Both strains showed increases in surface incongruity over time, as measured by increases in BSD. In B6 mice, acute surface incongruity was significantly correlated to the severity of PTA (R 2 = 0.988; p = .0006), but not in MRL mice (R 2 = 0.224; p = .220). PINP concentrations significantly decreased immediately post-fx in B6 mice (p = .023) but not in MRL mice, indicating higher bone synthesis in MRL mice. MRL/MpJ mice demonstrate a unique biologic response to articular fracture such that the observed articular bone surface displacement does not correlate with the severity of subsequent PTA. Clinical Relevance: Identifying therapies to enhance acute biologic repair following articular fracture may mitigate the risk of articular surface displacement for PTA.
Collapse
Affiliation(s)
- Tyler J Vovos
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Bridgette D Furman
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Janet L Huebner
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kelly A Kimmerling
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Gangadhar M Utturkar
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Cynthia L Green
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
| | - Virginia B Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA.,Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA.,Shriners Hospital for Children-St. Louis, St. Louis, Missouri, USA
| | - Steven A Olson
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
18
|
Glinkowski W, Chelnokov AN. The orthopaedic surgeon's clinical and experimental experience affect methods used for the fracture healing assessment (FHA) - An International Survey. Injury 2021; 52:2205-2214. [PMID: 33836844 DOI: 10.1016/j.injury.2021.03.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 02/02/2023]
Abstract
Detection of fracture healing (FH), which depends on assessment methods, is a crucial factor affecting treatment. The study aimed to examine orthopedic surgeons in terms of practical methods of fracture healing (FHA) assessment (physical, imaging, measurement, and laboratory) and to check whether surgeons participating in clinical and laboratory experiments or only clinical practitioners prefer different FHA methods. An International Survey on Fracture Healing Assessment Methods was developed and distributed through the Web-based survey portal. Ninety-three orthopedic surgeons, on average age 41.46 years, from 24 countries participated in the study. Thirty-one respondents (33.3%) reported dealing with fractures both in the clinic and in experimental studies, six (6.5%) reported dealing with fractures only in laboratory research work, and fifty-six (60.2%) indicated that they dealt with fractures only clinically. The survey's internal consistency was significantly high (Cronbach's alpha coefficients ranged from 0.84 to 0.96). The majority of respondents (80.83%) use specific clinical criteria to define a fracture union. The FHA was mainly based on the physical examination and plain radiograms. Laboratory findings, patient-oriented outcomes scores, and quantitative methods are rarely used. Orthopaedic surgeons dealing with fractures both in the clinic and in laboratory fracture research studies are more likely to use more quantitative FHA methods. Future research is needed to improve the international standard of the FHA methods for use in research, clinical trials, and clinical practice. Using a quantitative, reliable, and standardized approach, including online support, can be valuable for increasing compliance in the orthopedic surgeon population, effectively improving the adherence of fracture healing assessment in clinical conditions, and improving early detection of fracture healing disorders, improving fracture efficiency treatment.
Collapse
Affiliation(s)
- Wojciech Glinkowski
- Centre of Excellence "TeleOrto" for Telediagnostics and Treatment of Disorders and Injuries of the Locomotor System, Medical University of Warsaw, 00581 Warsaw, Poland; Department of Medical Informatics and Telemedicine, Medical University of Warsaw, 00581 Warsaw, Poland; Polish Telemedicine and eHealth Society, 03728 Warsaw, Poland; Gabinet Lekarski, 03728 Warsaw, Poland.
| | | |
Collapse
|
19
|
Hachemi Y, Rapp AE, Lee S, Dorn AK, Krüger BT, Kaiser K, Ignatius A, Tuckermann J. Intact Glucocorticoid Receptor Dimerization Is Deleterious in Trauma-Induced Impaired Fracture Healing. Front Immunol 2021; 11:628287. [PMID: 33679723 PMCID: PMC7927427 DOI: 10.3389/fimmu.2020.628287] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/29/2020] [Indexed: 11/13/2022] Open
Abstract
Following severe trauma, fracture healing is impaired because of overwhelming systemic and local inflammation. Glucocorticoids (GCs), acting via the glucocorticoid receptor (GR), influence fracture healing by modulating the trauma-induced immune response. GR dimerization-dependent gene regulation is essential for the anti-inflammatory effects of GCs. Therefore, we investigated in a murine trauma model of combined femur fracture and thoracic trauma, whether effective GR dimerization influences the pathomechanisms of trauma-induced compromised fracture healing. To this end, we used mice with decreased GR dimerization ability (GRdim). The healing process was analyzed by cytokine/chemokine multiplex analysis, flow cytometry, gene-expression analysis, histomorphometry, micro-computed tomography, and biomechanical testing. GRdim mice did not display a systemic or local hyper-inflammation upon combined fracture and thorax trauma. Strikingly, we discovered that GRdim mice were protected from fracture healing impairment induced by the additional thorax trauma. Collectively and in contrast to previous studies describing the beneficial effects of intact GR dimerization in inflammatory models, we report here an adverse role of intact GR dimerization in trauma-induced compromised fracture healing.
Collapse
Affiliation(s)
- Yasmine Hachemi
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Anna E Rapp
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Sooyeon Lee
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Ann-Kristin Dorn
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Benjamin T Krüger
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Kathrin Kaiser
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| |
Collapse
|
20
|
Hao Z, Li J, Li B, Alder KD, Cahill SV, Munger AM, Lee I, Kwon HK, Back J, Xu S, Kang MJ, Lee FY. Smoking Alters Inflammation and Skeletal Stem and Progenitor Cell Activity During Fracture Healing in Different Murine Strains. J Bone Miner Res 2021; 36:186-198. [PMID: 32866293 PMCID: PMC9057220 DOI: 10.1002/jbmr.4175] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/16/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
Abstract
Smokers are at a higher risk of delayed union or nonunion after fracture repair. Few specific interventions are available for prevention because the molecular mechanisms that result in these negative sequelae are poorly understood. Murine models that mimic fracture healing in smokers are crucial in further understanding the local cellular and molecular alterations during fracture healing caused by smoking. We exposed three murine strains, C57BL/6J, 129X1/SvJ, and BALB/cJ, to cigarette smoke for 3 months before the induction of a midshaft transverse femoral osteotomy. We evaluated fracture healing 4 weeks after the osteotomy using radiography, micro-computed tomography (μCT), and biomechanical testing. Radiographic analysis demonstrated a significant decrease in the fracture healing capacity of smoking 129X1/SvJ mice. μCT results showed delayed remodeling of fracture calluses in all three strains after cigarette smoke exposure. Biomechanical testing indicated the most significant impairment in the functional properties of 129X1/SvJ in comparison with C57BL/6J and BALB/cJ mice after cigarette smoke exposure. Thus, the 129X1/SvJ strain is most suitable in simulating smoking-induced impaired fracture healing. Furthermore, in smoking 129X1/SvJ murine models, we investigated the molecular and cellular alterations in fracture healing caused by cigarette smoking using histology, flow cytometry, and multiplex cytokine/chemokine analysis. Histological analysis showed impaired chondrogenesis in cigarette smoking. In addition, the important reparative cell populations, including skeletal stem cells and their downstream progenitors, demonstrated decreased expansion after injury as a result of cigarette smoking. Moreover, significantly increased pro-inflammatory mediators and the recruitment of immune cells in fracture hematomas were demonstrated in smoking mice. Collectively, our findings demonstrate the significant cellular and molecular alterations during fracture healing impaired by smoking, including disrupted chondrogenesis, aberrant skeletal stem and progenitor cell activity, and a pronounced initial inflammatory response. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Zichen Hao
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Emergency and Trauma, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jun Li
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Bo Li
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Orthopaedics, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Kareme D Alder
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Sean V Cahill
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Alana M Munger
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Inkyu Lee
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - JungHo Back
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Shuogui Xu
- Department of Emergency and Trauma, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Min-Jong Kang
- Department of Medicine, Pulmonary, Critical Care, and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| |
Collapse
|
21
|
Abstract
Our understanding of the mechanisms underlying fracture healing is rapidly developing and is contributing to new therapeutic strategies to enhance repair. To gain new insights, animal models must also evolve. From initially imprecise, uncontrolled bone defects we now have precise injury models that still capture all of the stages and phases of bone repair yet do so in a highly reproducible manner. The simple mono-cortical defect model allows assessment of bone repair through a cartilage intermediate, e.g., endochondral ossification, as well as direct bone repair, e.g., intramembranous healing. Cellular contributions of the periosteum can be distinguished from contributions originating in the bone marrow. In this chapter, we focus on the advantages of this bone repair model, as well as its limitations.
Collapse
Affiliation(s)
- Zhijun Li
- School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Jill A Helms
- School of Medicine, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
22
|
Moore-Lotridge SN, Ihejirika R, Gibson BHY, Posey SL, Mignemi NA, Cole HA, Hawley GD, Uppuganti S, Nyman JS, Schoenecker JG. Severe injury-induced osteoporosis and skeletal muscle mineralization: Are these related complications? Bone Rep 2020; 14:100743. [PMID: 33490313 PMCID: PMC7804603 DOI: 10.1016/j.bonr.2020.100743] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 12/10/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
Severely injured patients are beleaguered by complications during convalescence, such as dysregulated biomineralization. Paradoxically, severely injured patients experience the loss of bone (osteoporosis), resulting in diminished skeletal integrity and increased risk of fragility fractures; yet they also accrue mineralization in soft tissues, resulting in complications such as heterotopic ossification (HO). The pathophysiology leading to dysregulated biomineralization in severely injured patients is not well defined. It has been postulated that these pathologies are linked, such that mineralization is "transferred" from the bone to soft tissue compartments. The goal of this study was to determine if severe injury-induced osteoporosis and soft tissue calcification are temporally coincident following injury. Using a murine model of combined burn and skeletal muscle injury to model severe injury, it was determined that mice developed significant progressive bone loss, detectable as early as 3 days post injury, and marked soft tissue mineralization by 7 days after injury. The observed temporal concordance between the development of severe injury-induced osteoporosis and soft tissue mineralization indicates the plausibility that these complications share a common pathophysiology, though further experiments are required.
Collapse
Key Words
- BMD, bone mineral density
- BV/TV, bone volume/tissue volume
- Biomineralization
- Burn
- CTX, cardiotoxin
- DC, dystrophic calcification
- DPI, days post injury
- DXA, dual energy X-ray absorptiometry
- Dystrophic calcification
- H&E, hematoxylin and eosin
- HO, heterotopic ossification
- Heterotopic ossificaiton
- Osteoporosis
- STiCSS, soft tissue calcification scoring system
- Severe injury
- Severe injury-induced osteoporosis
- Soft tissue mineralization
- Trauma
- μCT, microcomputed tomography
Collapse
Affiliation(s)
- Stephanie N Moore-Lotridge
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Center of Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rivka Ihejirika
- Vanderbilt University Medical School, Vanderbilt University, Nashville, TN 37232, USA
| | - Breanne H Y Gibson
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Samuel L Posey
- Vanderbilt University Medical School, Vanderbilt University, Nashville, TN 37232, USA
| | - Nicholas A Mignemi
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Heather A Cole
- Department of Nuclear Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Gregory D Hawley
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sasidhar Uppuganti
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Center of Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Tennessee Valley Healthcare System, Vanderbilt University, Nashville, TN 37232, USA
| | - Jeffry S Nyman
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Center of Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Tennessee Valley Healthcare System, Vanderbilt University, Nashville, TN 37232, USA
| | - Jonathan G Schoenecker
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Center of Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
23
|
Bourzac C, Bensidhoum M, Manassero M, Chappard C, Michoux N, Pallu S, Portier H. Preventive Moderate Continuous Running-Exercise Conditioning Improves the Healing of Non-Critical Size Bone Defects in Male Wistar Rats: A Pilot Study Using µCT. Life (Basel) 2020; 10:life10120308. [PMID: 33255288 PMCID: PMC7760000 DOI: 10.3390/life10120308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/18/2022] Open
Abstract
Although physical exercise has unquestionable benefits on bone health, its effects on bone healing have been poorly investigated. This study evaluated the effects of preemptive moderate continuous running on the healing of non-critical sized bone defects in rats by µCT. We hypothesized that a preemptive running exercise would quicken bone healing. Twenty 5-week-old, male, Wistar rats were randomly allocated to one of the following groups (n = 10): sedentary control (SED) or continuous running (EX, 45 min/d, 5 d/week at moderate speed, for 8 consecutive weeks). A 2 mm diameter bone defect was then performed in the right tibia and femur. No exercise was performed during a 4 week-convalescence. Healing-tissue trabecular microarchitectural parameters were assessed once a week for 4 weeks using µCT and plasma bone turnover markers measured at the end of the study protocol (time point T12). At T12, bone volume fraction (BV/TV; BV: bone volume, TV: tissue volume) of the healing tissue in tibiae and femurs from EX rats was higher compared to that in SED rats (p = 0.001). BV/TV in EX rats was also higher in tibiae than in femurs (p < 0.01). The bone mineral density of the healing tissue in femurs from EX rats was higher compared to that in femurs from SED rats (p < 0.03). N-terminal telopeptide of collagen type I in EX rats was decreased compared to SED rats (p < 0.05), while no differences were observed for alkaline phosphatase and parathyroid hormone. The study provides evidence that preemptive moderate continuous running improves the healing of non-critical sized bone defects in male Wistar rats.
Collapse
Affiliation(s)
- Céline Bourzac
- B3OA, UMR CNRS 7052, INSERM U1271, Université de Paris, 75010 Paris, France; (C.B.); (M.B.); (M.M.); (C.C.); (S.P.)
- Département Elevage et Pathologie des Equidés et des Carnivores, Ecole Nationale Vétérinaire d’Alfort, 94700 Maisons-Alfort, France
| | - Morad Bensidhoum
- B3OA, UMR CNRS 7052, INSERM U1271, Université de Paris, 75010 Paris, France; (C.B.); (M.B.); (M.M.); (C.C.); (S.P.)
| | - Mathieu Manassero
- B3OA, UMR CNRS 7052, INSERM U1271, Université de Paris, 75010 Paris, France; (C.B.); (M.B.); (M.M.); (C.C.); (S.P.)
- Département Elevage et Pathologie des Equidés et des Carnivores, Ecole Nationale Vétérinaire d’Alfort, 94700 Maisons-Alfort, France
| | - Christine Chappard
- B3OA, UMR CNRS 7052, INSERM U1271, Université de Paris, 75010 Paris, France; (C.B.); (M.B.); (M.M.); (C.C.); (S.P.)
| | - Nicolas Michoux
- Département de Radiologie, Institut de Recherche Expérimentale et Clinique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Stéphane Pallu
- B3OA, UMR CNRS 7052, INSERM U1271, Université de Paris, 75010 Paris, France; (C.B.); (M.B.); (M.M.); (C.C.); (S.P.)
- Collegium Science & Technique, 2 Allée du Château, Université d’Orléans, 45100 Orléans, France
| | - Hugues Portier
- B3OA, UMR CNRS 7052, INSERM U1271, Université de Paris, 75010 Paris, France; (C.B.); (M.B.); (M.M.); (C.C.); (S.P.)
- Collegium Science & Technique, 2 Allée du Château, Université d’Orléans, 45100 Orléans, France
- Correspondence: ; Tel.: +33-782-309-43
| |
Collapse
|
24
|
Zhang F, Liu F, Yu S, Zhang G, Li J, Sun X. Protective Effect of Curcumin on Bone Trauma in a Rat Model via Expansion of Myeloid Derived Suppressor Cells. Med Sci Monit 2020; 26:e924724. [PMID: 33184252 PMCID: PMC7670833 DOI: 10.12659/msm.924724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Bone fracture, a common injury to bones leads to various biophysiological changes and pathological responses in the body. The current study investigated curcumin for treatment of bone fracture in a rat model of bone trauma, and evaluated the related mechanism. MATERIAL AND METHODS The rats were separated randomly into 3 groups; sham, model, and curcumin treatment groups. The fracture rat model was established by transverse osteotomy in the right femur bone at the mid-shaft. The osteoblast count was determined using hematoxylin and eosin staining. Vascular endothelial growth factor (VEGF) and proliferating cell nuclear antigen (PCNA) expression were measured by western blotting. RESULTS The rpS6-phosphorylation was suppressed and light chain 3 (LC3II) expression elevated in the curcumin treated group of the fracture rat model. In the curcumin-treated group, mineralization of fracture calluses was markedly higher on day 14 of fracture. The formation of osteoblasts was observed at a greater rate in the curcumin treated group compared to the model rat group. Treatment of rats with curcumin significantly (P<0.05) promoted expression of PCNA and VEGF. The decrease in CD11b+/Gr-1+ cell expansion in rats with bone trauma was alleviated significantly by curcumin treatment. A marked increase in arginase-1 expression in rats with bone trauma was caused by curcumin treatment. CONCLUSIONS In summary, curcumin activates autophagy and inhibits mTOR activation in bone tissues of rats with trauma. The curcumin promoted myeloid-derived suppressor cell (MDSC) proliferation and increased expansion of MDSCs in a rat model of trauma. Therefore, curcumin may have beneficial effect in patients with bone trauma and should be evaluated further for development of treatment.
Collapse
Affiliation(s)
- Futian Zhang
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Fu Liu
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Shaofen Yu
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Guihong Zhang
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Jie Li
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Xinjun Sun
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| |
Collapse
|
25
|
Hu CC, Chang CH, Chang Y, Hsieh JH, Ueng SWN. Beneficial Effect of TaON-Ag Nanocomposite Titanium on Antibacterial Capacity in Orthopedic Application. Int J Nanomedicine 2020; 15:7889-7900. [PMID: 33116507 PMCID: PMC7568682 DOI: 10.2147/ijn.s264303] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose In this study, a novel oxygenated nanocomposite thin film, TaON-Ag, was investigated in vitro and in vivo to evaluate its biocompatibility and antibacterial ability. Material and Methods The antibacterial ability of TaON-Ag nanocomposite-coated titanium (Ti) was evaluated using the Kirby-Bauer disk diffusion susceptibility test. The effects of TaON-Ag nanocomposite-coated metal on osteogenesis were further evaluated in an in vitro osteogenic culture model with rat marrow-derived mesenchymal stem cells (rMSCs). Furthermore, titanium rods coated with TaON-Ag were implanted into a rat femur fracture model either with or without osteomyelitis to investigate the effects of TaON-Ag in osteogenesis. Results The TaON-Ag-coated Ti exhibited an effective antibacterial effect against Staphylococcus aureus, coagulase-negative Staphylococcus, and the Gram-negative strains Escherichia coli and Pseudomonas aeruginosa. Using an osteogenic culture with rMSCs and a rat femoral fracture model, the TaON-Ag-coated Ti did not interfere with the ossification of rMSCs in vitro or during fracture healing in vivo. Field-emission scanning electron microscopy (FE-SEM) revealed that coating with TaON-Ag could inhibit pathogen adhesion and biofilm formation in both Staphylococcus aureus and Escherichia coli. Conclusion Using the proposed novel oxygenation process, TaON-Ag nanocomposite-coated Ti yielded robust biocompatibility and antibacterial ability against common microorganisms in orthopedic infections, thereby demonstrating potential for use in clinical applications.
Collapse
Affiliation(s)
- Chih-Chien Hu
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Kweishan, Taoyuan 33305, Taiwan.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Kweishan, Taoyuan 33305, Taiwan.,College of Medicine, Chang Gung University, Kweishan, Taoyuan 33302, Taiwan.,Department of Orthopedic Surgery, Xiamen Chang Gung Hospital, Xiamen, Fujian, People's Republic of China
| | - Chih-Hsiang Chang
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Kweishan, Taoyuan 33305, Taiwan.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Kweishan, Taoyuan 33305, Taiwan.,College of Medicine, Chang Gung University, Kweishan, Taoyuan 33302, Taiwan
| | - Yuhan Chang
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Kweishan, Taoyuan 33305, Taiwan.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Kweishan, Taoyuan 33305, Taiwan.,College of Medicine, Chang Gung University, Kweishan, Taoyuan 33302, Taiwan
| | - Jang-Hsing Hsieh
- Department of Materials Engineering, Ming Chi University of Technology, Taishan, Taipei 24301, Taiwan.,Center for Thin Film Technologies and Applications, Ming Chi University of Technology, Taishan, Taipei 24301, Taiwan
| | - Steve Wen-Neng Ueng
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Kweishan, Taoyuan 33305, Taiwan.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Kweishan, Taoyuan 33305, Taiwan.,College of Medicine, Chang Gung University, Kweishan, Taoyuan 33302, Taiwan
| |
Collapse
|
26
|
Administration of Human Non-Diabetic Mesenchymal Stromal Cells to a Murine Model of Diabetic Fracture Repair: A Pilot Study. Cells 2020; 9:cells9061394. [PMID: 32503335 PMCID: PMC7348854 DOI: 10.3390/cells9061394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 01/05/2023] Open
Abstract
Individuals living with type 1 diabetes mellitus may experience an increased risk of long bone fracture. These fractures are often slow to heal, resulting in delayed reunion or non-union. It is reasonable to theorize that the underlying cause of these diabetes-associated osteopathies is faulty repair dynamics as a result of compromised bone marrow progenitor cell function. Here it was hypothesized that the administration of non-diabetic, human adult bone marrow-derived mesenchymal stromal cells (MSCs) would enhance diabetic fracture healing. Human MSCs were locally introduced to femur fractures in streptozotocin-induced diabetic mice, and the quality of de novo bone was assessed eight weeks later. Biodistribution analysis demonstrated that the cells remained in situ for three days following administration. Bone bridging was evident in all animals. However, a large reparative callus was retained, indicating non-union. µCT analysis elucidated comparable callus dimensions, bone mineral density, bone volume/total volume, and volume of mature bone in all groups that received cells as compared to the saline-treated controls. Four-point bending evaluation of flexural strength, flexural modulus, and total energy to re-fracture did not indicate a statistically significant change as a result of cellular administration. An ex vivo lymphocytic proliferation recall assay indicated that the xenogeneic administration of human cells did not result in an immune response by the murine recipient. Due to this dataset, the administration of non-diabetic bone marrow-derived MSCs did not support fracture healing in this pilot study.
Collapse
|
27
|
Zieba J, Munivez E, Castellon A, Jiang MM, Dawson B, Ambrose CG, Lee B. Fracture Healing in Collagen-Related Preclinical Models of Osteogenesis Imperfecta. J Bone Miner Res 2020; 35:1132-1148. [PMID: 32053224 DOI: 10.1002/jbmr.3979] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/27/2020] [Accepted: 02/11/2020] [Indexed: 12/17/2022]
Abstract
Osteogenesis imperfecta (OI) is a genetic bone dysplasia characterized by bone deformities and fractures caused by low bone mass and impaired bone quality. OI is a genetically heterogeneous disorder that most commonly arises from dominant mutations in genes encoding type I collagen (COL1A1 and COL1A2). In addition, OI is recessively inherited with the majority of cases resulting from mutations in prolyl-3-hydroxylation complex members, which includes cartilage-associated protein (CRTAP). OI patients are at an increased risk of fracture throughout their lifetimes. However, non-union or delayed healing has been reported in 24% of fractures and 52% of osteotomies. Additionally, refractures typically go unreported, making the frequency of refractures in OI patients unknown. Thus, there is an unmet need to better understand the mechanisms by which OI affects fracture healing. Using an open tibial fracture model, our study demonstrates delayed healing in both Col1a2 G610c/+ and Crtap -/- OI mouse models (dominant and recessive OI, respectively) that is associated with reduced callus size and predicted strength. Callus cartilage distribution and chondrocyte maturation were altered in OI, suggesting accelerated cartilage differentiation. Importantly, we determined that healed fractured tibia in female OI mice are biomechanically weaker when compared with the contralateral unfractured bone, suggesting that abnormal OI fracture healing OI may prime future refracture at the same location. We have previously shown upregulated TGF-β signaling in OI and we confirm this in the context of fracture healing. Interestingly, treatment of Crtap -/- mice with the anti-TGF-β antibody 1D11 resulted in further reduced callus size and predicted strength, highlighting the importance of investigating dose response in treatment strategies. These data provide valuable insight into the effect of the extracellular matrix (ECM) on fracture healing, a poorly understood mechanism, and support the need for prevention of primary fractures to decrease incidence of refracture and deformity in OI patients. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jennifer Zieba
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Elda Munivez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Alexis Castellon
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brian Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Catherine G Ambrose
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
28
|
The association between mineralised tissue formation and the mechanical local in vivo environment: Time-lapsed quantification of a mouse defect healing model. Sci Rep 2020; 10:1100. [PMID: 31980656 PMCID: PMC6981157 DOI: 10.1038/s41598-020-57461-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 01/02/2020] [Indexed: 11/08/2022] Open
Abstract
An improved understanding of how local mechanical stimuli guide the fracture healing process has the potential to enhance clinical treatment of bone injury. Recent preclinical studies of bone defect in animal models have used cross-sectional data to examine this phenomenon indirectly. In this study, a direct time-lapsed imaging approach was used to investigate the local mechanical strains that precede the formation of mineralised tissue at the tissue scale. The goal was to test two hypotheses: 1) the local mechanical signal that precedes the onset of tissue mineralisation is higher in areas which mineralise, and 2) this local mechanical signal is independent of the magnitude of global mechanical loading of the tissue in the defect. Two groups of mice with femoral defects of length 0.85 mm (n = 10) and 1.45 mm (n = 9) were studied, allowing for distinct distributions of tissue scale strains in the defects. The regeneration and (re)modelling of mineralised tissue was observed weekly using in vivo micro-computed tomography (micro-CT), which served as a ground truth for resolving areas of mineralised tissue formation. The mechanical environment was determined using micro-finite element analysis (micro-FE) on baseline images. The formation of mineralised tissue showed strong association with areas of higher mechanical strain (area-under-the-curve: 0.91 ± 0.04, true positive rate: 0.85 ± 0.05) while surface based strains could correctly classify 43% of remodelling events. These findings support our hypotheses by showing a direct association between the local mechanical strains and the formation of mineralised tissue.
Collapse
|
29
|
PAZARCI Ö, DOĞAN HO, KILINÇ S, ÇAMURCU Y. Does mammalian target of rapamycin or sestrin 1 protein signaling have a role in bone
fracture healing? Turk J Med Sci 2019; 49:1774-1778. [PMID: 31655537 PMCID: PMC7518658 DOI: 10.3906/sag-1809-117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 09/22/2019] [Indexed: 11/30/2022] Open
Abstract
Background/aim Fracture healing is a complex physiological process that involves a well-orchestrated series of biological events. The mammalian target of rapamycin (mTOR) and sestrin 1 (SESN 1) play a central role in cell metabolism, proliferation, and survival. The aim of our study is to present serum mTOR and SESN 1 levels by comparing patients with or without bone fractures. It is also a guide for further research on the roles of these proteins in fracture healing. Materials and methods A total of 34 patients (10 females, 24 males) with bone fractures and 32 controls (10 females, 22 males) participated in this study. After collecting serum venous blood samples, the quantitative sandwich ELISA technique was used for the determination of serum mTOR and SESN 1 levels. Results The mean serum mTOR level was significantly higher in the fracture group compared to the control group (P = 0.001). However, SESN 1 levels did not significantly differ between groups (P = 0.913). Conclusion We found that serum mTOR levels increased on the first day after fracture compared to the control group. However, we obtained no significant difference between groups in terms of SESN 1 levels. This study may guide further clinical studies investigating the potential role of mTOR signaling in the bone healing process.
Collapse
Affiliation(s)
- Özhan PAZARCI
- Department of Orthopedics and Traumatology, Faculty of Medicine, Cumhuriyet University, SivasTurkey
- * To whom correspondence should be addressed. E-mail:
| | - Halef Okan DOĞAN
- Department of Biochemistry, Faculty of Science, Cumhuriyet University, SivasTurkey
| | - Seyran KILINÇ
- Department of Orthopedics and Traumatology, Faculty of Medicine, Cumhuriyet University, SivasTurkey
| | - Yalkin ÇAMURCU
- Department of Orthopedics and Traumatology, Faculty of Medicine, Erzincan University, ErzincanTurkey
| |
Collapse
|
30
|
Wehrle E, Tourolle Né Betts DC, Kuhn GA, Scheuren AC, Hofmann S, Müller R. Evaluation of longitudinal time-lapsed in vivo micro-CT for monitoring fracture healing in mouse femur defect models. Sci Rep 2019; 9:17445. [PMID: 31768003 PMCID: PMC6877534 DOI: 10.1038/s41598-019-53822-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/05/2019] [Indexed: 01/20/2023] Open
Abstract
Longitudinal in vivo micro-computed tomography (micro-CT) is of interest to non-invasively capture the healing process of individual animals in preclinical fracture healing studies. However, it is not known whether longitudinal imaging itself has an impact on callus formation and remodeling. In this study, a scan group received weekly micro-CT measurements (week 0-6), whereas controls were only scanned post-operatively and at week 5 and 6. Registration of consecutive scans using a branching scheme (bridged vs. unbridged defect) combined with a two-threshold approach enabled assessment of localized bone turnover and mineralization kinetics relevant for monitoring callus remodeling. Weekly micro-CT application did not significantly change any of the assessed callus parameters in the defect and periosteal volumes. This was supported by histomorphometry showing only small amounts of cartilage residuals in both groups, indicating progression towards the end of the healing period. Also, immunohistochemical staining of Sclerostin, previously associated with mediating adverse radiation effects on bone, did not reveal differences between groups. The established longitudinal in vivo micro-CT-based approach allows monitoring of healing phases in mouse femur defect models without significant effects of anesthesia, handling and radiation on callus properties. Therefore, this study supports application of longitudinal in vivo micro-CT for healing-phase-specific monitoring of fracture repair in mice.
Collapse
Affiliation(s)
- Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | - Sandra Hofmann
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
31
|
Goel PN, Moharrer Y, Hebb JH, Egol AJ, Kaur G, Hankenson KD, Ahn J, Ashley JW. Suppression of Notch Signaling in Osteoclasts Improves Bone Regeneration and Healing. J Orthop Res 2019; 37:2089-2103. [PMID: 31166033 PMCID: PMC6739141 DOI: 10.1002/jor.24384] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/28/2019] [Indexed: 02/04/2023]
Abstract
Owing to the central role of osteoclasts in bone physiology and remodeling, manipulation of their maturation process provides a potential therapeutic strategy for treating bone diseases. To investigate this, we genetically inhibited the Notch signaling pathway in the myeloid lineage, which includes osteoclast precursors, using a dominant negative form of MAML (dnMAML) that inhibits the transcriptional complex required for downstream Notch signaling. Osteoclasts derived from dnMAML mice showed no significant differences in early osteoclastic gene expression compared to the wild type. Further, these demonstrated significantly lowered resorption activity using bone surfaces while retaining their osteoblast stimulating ability using ex vivo techniques. Using in vivo approaches, we detected significantly higher bone formation rates and osteoblast gene expression in dnMAML cohorts. Further, these mice exhibited increased bone/tissue mineral density compared to wild type and larger bony calluses in later stages of fracture healing. These observations suggest that therapeutic suppression of osteoclast Notch signaling could reduce, but not eliminate, osteoclastic resorption without suppression of restorative bone remodeling and, therefore, presents a balanced paradigm for increasing bone formation, regeneration, and healing. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2089-2103, 2019.
Collapse
Affiliation(s)
- Peeyush N Goel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Yasaman Moharrer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - John H Hebb
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA,Georgetown University School of Medicine, Washington D.C
| | - Alexander J Egol
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | | | | | - Jaimo Ahn
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA,Co-corresponding Author: Jaimo Ahn, MD, PhD, FACS, FAOA, Advisory Dean, MSTP Steering Committee, Perelman School of Medicine, Co-Director, Orthopaedic Trauma, University of Pennsylvania Health System, Perelman School of Medicine, University of Pennsylvania, Investigator, Translational Musculoskeletal Research Center, Philadelphia Veterans Affairs Medical Center, 3737 Market Street, Floor 6, Philadelphia, PA-19104, Phone # +1 (215)-662-3340, Fax # +1 (215)-349-5890,
| | - Jason W Ashley
- Eastern Washington University, Cheney, WA,Corresponding Author: Jason Waid Ashley, PhD, Assistant Professor, Biology Department, 526 5th Street, SCI236, Eastern Washington University, Cheney, WA 99004, Phone # +1(509)-359-4665,
| |
Collapse
|
32
|
The Size of Intramedullary Fixation Affects Endochondral-Mediated Angiogenesis During Fracture Repair. J Orthop Trauma 2019; 33:e385-e393. [PMID: 31259800 DOI: 10.1097/bot.0000000000001555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES To explore the effect of intramedullary pin size on the biology of a healing fracture, specifically endochondral angiogenesis. We hypothesized that fracture fixation with a smaller pin would permit greater interfragmentary strain resulting in increased total amount of vascular endothelial growth factor within the callus and greater angiogenesis compared to fixation with a larger pin. METHODS Transverse mid-shaft femur fractures in 8-week-old mice were fixed with either a 23-gauge (G) or 30-G pin. Differences in interfragmentary strain at the fracture site were estimated between cohorts. A combination of histology, gene expression, serial radiography, and microcomputed tomography with and without vascular contrast agent were used to assess fracture healing and vascularity for each cohort. RESULTS Larger soft-tissue callus formation increased vascular endothelial growth factor-A expression, and a corresponding increase in vascular volume was observed in the higher strain, 30-G cohort. Radiographic analysis demonstrated earlier hard callus formation with greater initial interfragmentary strain, similar rates of union between pin size cohorts, yet delayed callus remodeling in mice with the larger pin size. CONCLUSIONS These findings suggest that the stability conferred by an intramedullary nail influences endochondral angiogenesis at the fracture.
Collapse
|
33
|
Orth M, Baudach J, Scheuer C, Osche D, Veith N, Braun B, Rollmann M, Herath S, Pohlemann T, Menger M, Histing T. Erythropoietin does not improve fracture healing in aged mice. Exp Gerontol 2019; 122:1-9. [DOI: 10.1016/j.exger.2019.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/05/2019] [Accepted: 04/12/2019] [Indexed: 01/08/2023]
|
34
|
Adipose-derived stem cells prevent the onset of bisphosphonate-related osteonecrosis of the jaw through transforming growth factor β-1-mediated gingival wound healing. Stem Cell Res Ther 2019; 10:169. [PMID: 31196208 PMCID: PMC6567445 DOI: 10.1186/s13287-019-1277-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/27/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022] Open
Abstract
Background Due to its complex pathogenesis and low clinical cure rate, bisphosphonate-related osteonecrosis of the jaw (BRONJ) poses a substantial challenge for oral and maxillofacial surgeons. Therefore, the treatment of BRONJ should focus on prevention. In clinical studies, primary wound closure can significantly reduce the incidence of BRONJ. Whether local stem cell transplantation can promote primary gingival healing in patients with a medication history and prevent BRONJ has not been reported. Methods In this study, animals were divided into a healthy group (non-drug treatment), a BP group, a hydroxyapatite (HA) group, and an adipose-derived stem cell (ADSC) group. All groups except the healthy group were treated with BPs and immunosuppressive drugs once per week for 8 weeks, simulating clinical use for the treatment of cancer patients with bone metastasis, to induce BRONJ-like animals. After the sixth drug treatment, the bilateral premolars were extracted in all groups. In contrast to the healthy and BP groups, the extraction sockets in the HA and ADSC groups were filled with HA or HA + ADSCs simultaneously post extraction to observe the preventive effect of ADSCs on the occurrence of BRONJ. At 2 and 8 weeks post extraction, animals from all groups were sacrificed. Results At 8 weeks post transplantation, ADSCs prevented the occurrence of BRONJ, mainly through accelerating healing of the gingival epithelium at 2 weeks post extraction. We also found that ADSCs could upregulate the expression of transforming growth factor β1 (TGF-β1) and fibronectin in tissue from animals with a medication history by accelerating gingival healing of the extraction socket. A rescue assay further demonstrated that TGF-β1 and fibronectin expression decreased in TGF-β1-deficient ADSC-treated animals, which partially abolished the preventive effect of ADSCs on the onset of BRONJ. Conclusion ADSCs prevent the onset of BRONJ, mainly by upregulating the expression of TGF-β1 and fibronectin to promote primary gingival healing, ultimately leading to bone regeneration in the tooth extraction socket. Our new findings provide a novel stem cell treatment for the prevention of BRONJ. Electronic supplementary material The online version of this article (10.1186/s13287-019-1277-y) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Chen MQ, Luan JJ. HMGB1 promotes bone fracture healing through activation of ERK signaling pathway in a rat tibial fracture model. Kaohsiung J Med Sci 2019; 35:550-558. [PMID: 31162822 DOI: 10.1002/kjm2.12095] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 05/12/2019] [Indexed: 01/04/2023] Open
Abstract
This work was to investigate potential roles of HMGB1-mediated ERK pathway in the healing process of bone fracture. Rat tibial fracture models were established and divided into control (rats with normal saline), HMGB1 (rats with HMGB1), and HMGB1+ PD98059 groups (rats with HMGB1 and 1 mg/kg of ERK1/2 inhibitor PD98059) with 30 rats per each. The healing of rats' fracture was observed by X-ray films, the morphological changes of bone fractures by HE staining, the callus formation by micro-CT and biomechanical test, and the expression of osteogenesis-related genes, HMGB1 and ERK-related proteins by qRT-PCR and Western blot. Rats in the HMGB1 group was increased in X-ray scores, peak torque, torsional stiffness, and the bone volume fraction (bone volume/total volume, BV/TV); meanwhile, those rats presented elevations in osteogenesis-related genes and HMGB1 expressions, as well as p-ERK/ERK ratio. However, rats in the HMGB1+ PD98059 group was significantly reduced in X-ray score, peak torque, torsional stiffness, and BV/TV, as well as the expression of osteogenesis-related genes and the ratio of p-ERK/ERK, as compared to those from HMGB1 group. HMGB1 could promote the expressions of osteogenesis-related genes and accelerate the healing process of fracture via activation of the ERK signaling pathway.
Collapse
Affiliation(s)
- Ming-Qi Chen
- Department of Traumatic Orthopedics, YanTaiShan Hospital, YanTai City, Shandong, China
| | - Jing-Jie Luan
- Department of Traumatic Orthopedics, YanTaiShan Hospital, YanTai City, Shandong, China
| |
Collapse
|
36
|
Lin MC, Hu D, Marmor M, Herfat ST, Bahney CS, Maharbiz MM. Smart bone plates can monitor fracture healing. Sci Rep 2019; 9:2122. [PMID: 30765721 PMCID: PMC6375940 DOI: 10.1038/s41598-018-37784-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/07/2018] [Indexed: 11/08/2022] Open
Abstract
There are currently no standardized methods for assessing fracture healing, with physicians relying on X-rays which are only useful at later stages of repair. Using in vivo mouse fracture models, we present the first evidence that microscale instrumented implants provide a route for post-operative fracture monitoring, utilizing electrical impedance spectroscopy (EIS) to track the healing tissue with high sensitivity. In this study, we fixed mouse long bone fractures with external fixators and bone plates. EIS measurements taken across two microelectrodes within the fracture gap were able to track longitudinal differences between individual mice with good versus poor healing. We additionally present an equivalent circuit model that combines the EIS data to classify fracture repair states. Lastly, we show that EIS measurements strongly correlated with standard quantitative µCT values and that these correlations validate clinically-relevant operating frequencies for implementation of this technique. These results demonstrate that EIS can be integrated into current fracture management strategies such as bone plating, providing physicians with quantitative information about the state of fracture repair to guide clinical decision-making for patients.
Collapse
Affiliation(s)
- Monica C Lin
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA.
| | - Diane Hu
- UCSF Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, 94110, USA
| | - Meir Marmor
- UCSF Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, 94110, USA
| | - Safa T Herfat
- UCSF Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, 94110, USA
| | - Chelsea S Bahney
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
- UCSF Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, 94110, USA
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, 81657, USA
| | - Michel M Maharbiz
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
| |
Collapse
|
37
|
Qiao J, Zhou M, Li Z, Ren J, Gao G, Cao G, Shen H, Lu S. Comparison of remote ischemic preconditioning and intermittent hypoxia training in fracture healing. Mol Med Rep 2018; 19:1867-1874. [PMID: 30592272 DOI: 10.3892/mmr.2018.9788] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 11/07/2018] [Indexed: 11/06/2022] Open
Abstract
Fracture healing in elderly patients is an emerging public health concern. As non‑drug treatments, intermittent hypoxia training (IHT) and remote ischemic preconditioning (RIPC) are considered to have substantial advantages and to aid fracture healing in elderly patients. The purpose of the present study was to evaluate and compare the effects of IHT and RIPC on fracture healing. Micro‑computed tomography (micro‑CT) and biomechanical testing were used to assess the morphology and structural properties of bone callus dissected from aged rats with tibial fractures. In addition, hypoxia‑inducible factor‑1α (HIF‑1α) and its target gene, associated with the healing process, were investigated by reverse transcription‑quantitative polymerase chain reaction and western blot analyses. The micro‑CT‑based parameters, including bone mineral density and trabecular number, were measured, and significant differences were identified between the experimental and control groups. The IHT group exhibited superior bone formation and mineralization rates compared with the RIPC group. The biomechanical testing revealed that the ultimate loading and stiffness values were significantly higher in the IHT group compared with those in the RIPC group. In accordance with previous studies, RIPC exerted a similar effect in increasing the expression of HIF‑1α, and its downstream genes, throughout the course of healing. In addition, the IHT group exhibited increased expression levels of HIF‑1α compared with the RIPC group. Taken together, the results suggested that IHT and RIPC significantly enhanced fracture healing; however, IHT exhibited superior bone formation and healing effects compared with RIPC.
Collapse
Affiliation(s)
- Junjie Qiao
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Meng Zhou
- Department of Orthopedics, Beijing Jishuitan Hospital, 4th Medical College of Peking University, Beijing 100035, P.R. China
| | - Zheng Li
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Jie Ren
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Guanghan Gao
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Guanglei Cao
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Huiliang Shen
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Shibao Lu
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| |
Collapse
|
38
|
Bravo D, Josephson AM, Bradaschia-Correa V, Wong MZ, Yim NL, Neibart SS, Lee SN, Huo J, Coughlin T, Mizrahi MM, Leucht P. Temporary inhibition of the plasminogen activator inhibits periosteal chondrogenesis and promotes periosteal osteogenesis during appendicular bone fracture healing. Bone 2018; 112:97-106. [PMID: 29680264 PMCID: PMC5970081 DOI: 10.1016/j.bone.2018.04.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 02/11/2018] [Accepted: 04/17/2018] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Aminocaproic acid is approved as an anti-fibrinolytic for use in joint replacement and spinal fusion surgeries to limit perioperative blood loss. Previous animal studies have demonstrated a pro-osteogenic effect of aminocaproic acid in spine fusion models. Here, we tested if aminocaproic acid enhances appendicular bone healing and we sought to uncover the effect of aminocaproic acid on osteoprogenitor cells (OPCs) during bone regeneration. METHODS We employed a well-established murine femur fracture model in adult C57BL/6J mice after receiving two peri-operative injections of aminocaproic acid. Routine histological assays, biomechanical testing and micro-CT analyses were utilized to assess callus volume, and strength, progenitor cell proliferation, differentiation, and remodeling in vivo. Two disparate ectopic transplantation models were used to study the effect of the growth factor milieu within the early fracture hematoma on osteoprogenitor cell fate decisions. RESULTS Aminocaproic acid treated femur fractures healed with a significantly smaller cartilaginous callus, and this effect was also observed in the ectopic transplantation assays. We hypothesized that aminocaproic acid treatment resulted in a stabilization of the early fracture hematoma, leading to a change in the growth factor milieu created by the early hematoma. Gene and protein expression analysis confirmed that aminocaproic acid treatment resulted in an increase in Wnt and BMP signaling and a decrease in TGF-β-signaling, resulting in a shift from chondrogenic to osteogenic differentiation in this model of endochondral bone formation. CONCLUSION These experiments demonstrate for the first time that inhibition of the plasminogen activator during fracture healing using aminocaproic acid leads to a change in cell fate decision of periosteal osteoprogenitor cells, with a predominance of osteogenic differentiation, resulting in a larger and stronger bony callus. These findings may offer a promising new use of aminocaproic acid, which is already FDA-approved and offers a very safe risk profile.
Collapse
Affiliation(s)
- D Bravo
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - A M Josephson
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - V Bradaschia-Correa
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - M Z Wong
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - N L Yim
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - S S Neibart
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - S N Lee
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - J Huo
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - T Coughlin
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - M M Mizrahi
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - P Leucht
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States.
| |
Collapse
|
39
|
Yuasa M, Saito M, Molina C, Moore-Lotridge SN, Benvenuti MA, Mignemi NA, Okawa A, Yoshii T, Schwartz HS, Nyman JS, Schoenecker JG. Unexpected timely fracture union in matrix metalloproteinase 9 deficient mice. PLoS One 2018; 13:e0198088. [PMID: 29851987 PMCID: PMC5978876 DOI: 10.1371/journal.pone.0198088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/14/2018] [Indexed: 11/18/2022] Open
Abstract
Immediately following a fracture, a fibrin laden hematoma is formed to prevent bleeding and infection. Subsequently, the organized removal of fibrin, via the protease plasmin, is essential to permit fracture repair through angiogenesis and ossification. Yet, when plasmin activity is lost, the depletion of fibrin alone is insufficient to fully restore fracture repair, suggesting the existence of additional plasmin targets important for fracture repair. Previously, activated matrix metalloproteinase 9 (MMP-9) was demonstrated to function in fracture repair by promoting angiogenesis. Given that MMP-9 is a defined plasmin target, it was hypothesized that pro-MMP-9, following plasmin activation, promotes fracture repair. This hypothesis was tested in a fixed murine femur fracture model with serial assessment of fracture healing. Contrary to previous findings, a complete loss of MMP-9 failed to affect fracture healing and union through 28 days post injury. Therefore, these results demonstrated that MMP-9 is dispensable for timely fracture union and cartilage transition to bone in fixed femur fractures. Pro-MMP-9 is therefore not a significant target of plasmin in fracture repair and future studies assessing additional plasmin targets associated with angiogenesis are warranted.
Collapse
Affiliation(s)
- Masato Yuasa
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masanori Saito
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Cesar Molina
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Stephanie N. Moore-Lotridge
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Michael A. Benvenuti
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Nicholas A. Mignemi
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Atsushi Okawa
- Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshitaka Yoshii
- Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Herbert S. Schwartz
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jeffry S. Nyman
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, Tennessee, United States of America
| | - Jonathan G. Schoenecker
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
40
|
Natoli RM, Yu H, Meislin MCM, Abbasnia P, Roper P, Vuchkovska A, Xiao X, Stock SR, Callaci JJ. Alcohol exposure decreases osteopontin expression during fracture healing and osteopontin-mediated mesenchymal stem cell migration in vitro. J Orthop Surg Res 2018; 13:101. [PMID: 29699560 PMCID: PMC5921778 DOI: 10.1186/s13018-018-0800-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 04/03/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Alcohol consumption is a risk factor for impaired fracture healing, though the mechanism(s) by which this occurs are not well understood. Our laboratory has previously shown that episodic alcohol exposure of rodents negatively affects fracture callus development, callus biomechanics, and cellular signaling which regulates stem cell differentiation. Here, we examine whether alcohol alters chemokine expression and/or signaling activity in the mouse fracture callus during early fracture healing. METHODS A mouse model for alcohol-impaired tibia fracture healing was utilized. Early fracture callus was examined for alcohol-effects on tissue composition, expression of chemokines involved in MSC migration to the fracture site, and biomechanics. The effects of alcohol on MSC migration and cell adhesion receptors were examined in an in vitro system. RESULTS Mice exposed to alcohol showed decreased evidence of external callus formation, decreased callus-related osteopontin (OPN) expression levels, and decreased biomechanical stiffness. Alcohol exposure decreased rOPN-mediated MSC migration and integrin β1 receptor expression in vitro. CONCLUSIONS The effects of alcohol exposure demonstrated here on fracture callus-associated OPN expression, rOPN-mediated MSC migration in vitro, and MSC integrin β1 receptor expression in vitro have not been previously reported. Understanding the effects of alcohol exposure on the early stages of fracture repair may allow timely initiation of treatment to mitigate the long-term complications of delayed healing and/or fracture non-union.
Collapse
Affiliation(s)
- Roman M Natoli
- Department of Orthopaedic Surgery and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, 2160 South First Ave, Maywood, IL, 60153, USA.,Present Address: Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Henry Yu
- Department of Orthopaedic Surgery and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, 2160 South First Ave, Maywood, IL, 60153, USA
| | - Megan Conti-Mica Meislin
- Department of Orthopaedic Surgery and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, 2160 South First Ave, Maywood, IL, 60153, USA.,Present Address: Department of Orthopaedic Surgery and Rehabilitation Medicine, Hand and Upper Extremity Division, The University of Chicago, Chicago, IL, USA
| | - Pegah Abbasnia
- Department of Orthopaedic Surgery and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, 2160 South First Ave, Maywood, IL, 60153, USA.,Present Address: School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip Roper
- Department of Orthopaedic Surgery and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, 2160 South First Ave, Maywood, IL, 60153, USA
| | - Aleksandra Vuchkovska
- Department of Orthopaedic Surgery and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, 2160 South First Ave, Maywood, IL, 60153, USA
| | - Xianghui Xiao
- Present Address: Argonne National Laboratory Advanced Photon Source, Lemont, IL, USA
| | - Stuart R Stock
- Present Address: School of Medicine, Northwestern University Feinberg, Chicago, IL, USA
| | - John J Callaci
- Department of Orthopaedic Surgery and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, 2160 South First Ave, Maywood, IL, 60153, USA.
| |
Collapse
|
41
|
Fully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to G Alert. Proc Natl Acad Sci U S A 2018; 115:E4463-E4472. [PMID: 29674451 PMCID: PMC5949009 DOI: 10.1073/pnas.1802893115] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
While stem cell therapy has become the standard of care for hematological disorders, challenges remain for the treatment of solid organ injuries. Targeting endogenous cells would overcome many hurdles associated with exogenous stem cell therapy. Alarmins are released upon tissue damage, and here we describe how upregulation of a physiological pathway by exogenous administration of a single dose of HMGB1, either locally or systemically, promotes tissue repair by targeting endogenous stem cells. We show that HMGB1 complexed with CXCL12 transitions stem cells that express CXCR4 from G0 to GAlert. These primed cells rapidly respond to appropriate activating factors released upon injury. HMGB1 promotes healing even if administered 2 wk before injury, thereby expanding its translational benefit for diverse clinical scenarios. A major discovery of recent decades has been the existence of stem cells and their potential to repair many, if not most, tissues. With the aging population, many attempts have been made to use exogenous stem cells to promote tissue repair, so far with limited success. An alternative approach, which may be more effective and far less costly, is to promote tissue regeneration by targeting endogenous stem cells. However, ways of enhancing endogenous stem cell function remain poorly defined. Injury leads to the release of danger signals which are known to modulate the immune response, but their role in stem cell-mediated repair in vivo remains to be clarified. Here we show that high mobility group box 1 (HMGB1) is released following fracture in both humans and mice, forms a heterocomplex with CXCL12, and acts via CXCR4 to accelerate skeletal, hematopoietic, and muscle regeneration in vivo. Pretreatment with HMGB1 2 wk before injury also accelerated tissue regeneration, indicating an acquired proregenerative signature. HMGB1 led to sustained increase in cell cycling in vivo, and using Hmgb1−/− mice we identified the underlying mechanism as the transition of multiple quiescent stem cells from G0 to GAlert. HMGB1 also transitions human stem and progenitor cells to GAlert. Therefore, exogenous HMGB1 may benefit patients in many clinical scenarios, including trauma, chemotherapy, and elective surgery.
Collapse
|
42
|
Schindeler A, Mills RJ, Bobyn JD, Little DG. Preclinical models for orthopedic research and bone tissue engineering. J Orthop Res 2018; 36:832-840. [PMID: 29205478 DOI: 10.1002/jor.23824] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/27/2017] [Indexed: 02/04/2023]
Abstract
In this review, we broadly define and discuss the preclinical rodent models that are used for orthopedics and bone tissue engineering. These range from implantation models typically used for biocompatibility testing and high-throughput drug screening, through to fracture and critical defect models used to model bone healing and severe orthopedic injuries. As well as highlighting the key methods papers describing these techniques, we provide additional commentary based on our substantive practical experience with animal surgery and in vivo experimental design. This review also briefly touches upon the descriptive and functional outcome measures and power calculations that are necessary for an informative study. Obtaining informative and relevant research outcomes can be very dependent on the model used, and we hope this evaluation of common models will serve as a primer for new researchers looking to undertake preclinical bone studies. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:832-840, 2018.
Collapse
Affiliation(s)
- Aaron Schindeler
- Orthopedic Research and Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, Sydney, New South Wales, 2145, Australia.,Discipline of Pediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Rebecca J Mills
- Orthopedic Research and Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, Sydney, New South Wales, 2145, Australia
| | - Justin D Bobyn
- Orthopedic Research and Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, Sydney, New South Wales, 2145, Australia.,Discipline of Pediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - David G Little
- Orthopedic Research and Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, Sydney, New South Wales, 2145, Australia.,Discipline of Pediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| |
Collapse
|
43
|
Cooke ME, Hussein AI, Lybrand KE, Wulff A, Simmons E, Choi JH, Litrenta J, Ricci WM, Nascone JW, O’Toole RV, Morgan EF, Gerstenfeld LC, Tornetta P. Correlation between RUST assessments of fracture healing to structural and biomechanical properties. J Orthop Res 2018; 36:945-953. [PMID: 28833572 PMCID: PMC5823715 DOI: 10.1002/jor.23710] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/19/2017] [Indexed: 02/04/2023]
Abstract
Radiographic Union Score for Tibia (RUST) and modified RUST (mRUST) are radiographic tools for quantitatively evaluating fracture healing using a cortical scoring system. This tool has high intra-class correlation coefficients (ICCs); however, little evidence has evaluated the scores against the physical properties of bone healing. Closed, stabilized fractures were made in the femora of C3H/HeJ male mice (8-12 week-old) of two dietary groups: A control and a phosphate restricted diet group. Micro-computed tomography (µCT) and torsion testing were carried out at post-operative days (POD) 14, 21, 35, and 42 (n = 10-16) per group time-point. Anteroposterior and lateral radiographic views were constructed from the µCT scans and scored by five raters. The raters also indicated if the fracture were healed. ICCs were 0.71 (mRUST) and 0.63 (RUST). Both RUST scores were positively correlated with callus bone mineral density (BMD) (r = 0.85 and 0.80, p < 0.001) and bone volume fraction (BV/TV) (r = 0.86 and 0.80, p < 0.001). Both RUST scores positively correlated with callus strength (r = 0.35 and 0.26, p < 0.012) and rigidity (r = 0.50 and 0.39, p < 0.001). Radiographically healed calluses had a mRUST ≥13 and a RUST ≥10 and had excellent relationship to structural and biomechanical metrics. Effect of delayed healing due to phosphate dietary restrictions was found at later time points with all mechanical properties (p < 0.011), however no differences found in the RUST scores (p > 0.318). Clinical relevance of this study is both RUST scores showed high correlation to physical properties of healing and generally distinguished healed vs. non-healed fractures. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:945-953, 2018.
Collapse
Affiliation(s)
- Margaret E Cooke
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Amira I Hussein
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Kyle E Lybrand
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Alexander Wulff
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Erin Simmons
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Jeffrey H Choi
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Jody Litrenta
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - William M Ricci
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO
| | - Jason W Nascone
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Robert V O’Toole
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Elise F Morgan
- Department of Mechanical Engineering, Boston University, Boston, MA
| | | | - Paul Tornetta
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| |
Collapse
|
44
|
Li C, Tan R, Guo Y, Li S. Using 3D finite element models verified the importance of callus material and microstructure in biomechanics restoration during bone defect repair. Comput Methods Biomech Biomed Engin 2018; 21:83-90. [PMID: 29359598 DOI: 10.1080/10255842.2018.1425404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Chentian Li
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Rongwei Tan
- Research and Development Center, Shenzhen Lando Biomaterial Co.Ltd., Shenzhen, China
| | - Yuanjun Guo
- Research and Development Center, Shenzhen Lando Biomaterial Co.Ltd., Shenzhen, China
| | - Songjian Li
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
45
|
Bradaschia-Correa V, Josephson AM, Egol AJ, Mizrahi MM, Leclerc K, Huo J, Cronstein BN, Leucht P. Ecto-5'-nucleotidase (CD73) regulates bone formation and remodeling during intramembranous bone repair in aging mice. Tissue Cell 2017; 49:545-551. [PMID: 28720305 PMCID: PMC5656528 DOI: 10.1016/j.tice.2017.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 07/03/2017] [Accepted: 07/03/2017] [Indexed: 11/19/2022]
Abstract
Ecto-5'-nucleotidase (CD73) generates adenosine, an osteoblast activator and key regulator of skeletal growth. It is unknown, however, if CD73 regulates osteogenic differentiation during fracture healing in adulthood, and in particular how CD73 activity regulates intramembranous bone repair in the elderly. Monocortical tibial defects were created in 46-52-week-old wild type (WT) and CD73 knock-out mice (CD73-/-) mice. Injury repair was analyzed at post-operative days 5, 7, 14 and 21 by micro-computed tomography (micro-CT), histomorphometry, proliferating cell nuclear antigen (PCNA) immunostaining, alkaline phosphatase (ALP) and tartrate-resistant acid phosphatase (TRAP) histochemistry. Middle-aged CD73 knock-out mice exhibited delayed bone regeneration and significantly reduced bone matrix deposition detected by histomorphometry and micro-CT. Cell proliferation, ALP activity and osteoclast number were reduced in the CD73-/- mice, suggesting a combined defect in bone formation and resorption due the absence of CD73 activity in this model of intramembranous bone repair. Results from this study demonstrate that osteoblast activation through CD73 activity is essential during bone repair in aging mice, and it may present a drugable target for future biomimetic therapeutic approaches that aim at enhancing bone formation in the elderly patients.
Collapse
Affiliation(s)
- Vivian Bradaschia-Correa
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Anne M Josephson
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Alexander J Egol
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Matthew M Mizrahi
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Kevin Leclerc
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Jason Huo
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA
| | - Bruce N Cronstein
- Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Philipp Leucht
- Department of Orthopaedic Surgery, New York University Langone Medical Center - Hospital for Joint Diseases, New York, NY, USA; Department of Cell Biology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
46
|
Wang P, Ying J, Luo C, Jin X, Zhang S, Xu T, Zhang L, Mi M, Chen D, Tong P, Jin H. Osthole Promotes Bone Fracture Healing through Activation of BMP Signaling in Chondrocytes. Int J Biol Sci 2017; 13:996-1007. [PMID: 28924381 PMCID: PMC5599905 DOI: 10.7150/ijbs.19986] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/18/2017] [Indexed: 11/16/2022] Open
Abstract
Osthole is a bioactive coumarin derivative and has been reported to be able to enhance bone formation and improve fracture healing. However, the molecular mechanism of Osthole in bone fracture healing has not been fully defined. In this study we determined if Osthole enhances bone fracture healing through activation of BMP2 signaling in mice. We performed unilateral open transverse tibial fracture procedure in 10-week-old C57BL/6 mice which were treated with or without Osthole. Our previous studies demonstrated that chondrocyte BMP signaling is required for bone fracture healing, in this study we also performed tibial fracture procedure in Cre-negative and Col2-Cre;Bmp2flox/flox conditional knockout (KO) mice (Bmp2Col2Cre) to determine if Osthole enhances fracture healing in a BMP2-dependent manner. Fracture callus tissues were collected and analyzed by X-ray, micro-CT (μCT), histology, histomorphometry, immunohistochemistry (IHC), biomechanical testing and quantitative gene expression analysis. In addition, mouse chondrogenic ATDC5 cells were cultured with or without Osthole and the expression levels of chondrogenic marker genes were examined. The results demonstrated that Osthole promotes bone fracture healing in wild-type (WT) or Cre- control mice. In contrast, Osthole failed to promote bone fracture healing in Bmp2Col2Creconditional KO mice. In the mice receiving Osthole treatment, expression of cartilage marker genes was significantly increased. We conclude that Osthole could promote bone strength and enhance fracture healing by activation of BMP2 signaling. Osthole may be used as an alternative approach in the orthopaedic clinic for the treatment of fracture healing.
Collapse
Affiliation(s)
- Pinger Wang
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jun Ying
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,First Clinical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Cheng Luo
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,First Clinical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Xing Jin
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,Department of Orthopaedics and Traumatology, Wangjiang Sub-District Community Health Service Centre, Hangzhou 310016, Zhejiang Province, China
| | - Shanxing Zhang
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Taotao Xu
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,First Clinical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Lei Zhang
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,First Clinical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Meng Mi
- Department of Traumatology, Beijing Jishuitan Hospital, 100035, Beijing, China
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - Peijian Tong
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
47
|
Bradaschia-Correa V, Josephson AM, Mehta D, Mizrahi M, Neibart SS, Liu C, Kennedy OD, Castillo AB, Egol KA, Leucht P. The Selective Serotonin Reuptake Inhibitor Fluoxetine Directly Inhibits Osteoblast Differentiation and Mineralization During Fracture Healing in Mice. J Bone Miner Res 2017; 32:821-833. [PMID: 27869327 PMCID: PMC5395314 DOI: 10.1002/jbmr.3045] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 11/10/2022]
Abstract
Chronic use of selective serotonin reuptake inhibitors (SSRIs) for the treatment of depression has been linked to osteoporosis. In this study, we investigated the effect of chronic SSRI use on fracture healing in two murine models of bone regeneration. First, we performed a comprehensive analysis of endochondral bone healing in a femur fracture model. C57/BL6 mice treated with fluoxetine, the most commonly prescribed SSRI, developed a normal cartilaginous soft-callus at 14 days after fracture and demonstrated a significantly smaller and biomechanically weaker bony hard-callus at 28 days. In order to further dissect the mechanism that resulted in a smaller bony regenerate, we used an intramembranous model of bone healing and revealed that fluoxetine treatment resulted in a significantly smaller bony callus at 7 and 14 days postinjury. In order to test whether the smaller bony regenerate following fluoxetine treatment was caused by an inhibition of osteogenic differentiation and/or mineralization, we employed in vitro experiments, which established that fluoxetine treatment decreases osteogenic differentiation and mineralization and that this effect is serotonin-independent. Finally, in a translational approach, we tested whether cessation of the medication would result in restoration of the regenerative potential. However, histologic and μCT analysis revealed non-union formation in these animals with fibrous tissue interposition within the callus. In conclusion, fluoxetine exerts a direct, inhibitory effect on osteoblast differentiation and mineralization, shown in two disparate murine models of bone repair. Discontinuation of the drug did not result in restoration of the healing potential, but rather led to complete arrest of the repair process. Besides the well-established effect of SSRIs on bone homeostasis, our study provides strong evidence that fluoxetine use negatively impacts fracture healing. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Vivian Bradaschia-Correa
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA
| | - Anne M Josephson
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA
| | - Devan Mehta
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA
| | - Matthew Mizrahi
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA
| | - Shane S Neibart
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA
| | - Chao Liu
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA.,Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, New York, NY, USA
| | - Oran D Kennedy
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA.,Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, New York, NY, USA
| | - Alesha B Castillo
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA.,Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, New York, NY, USA
| | - Kenneth A Egol
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA
| | - Philipp Leucht
- Department of Orthopaedic Surgery, Langone Medical Center-Hospital for Joint Diseases, New York University, New York, NY, USA.,Department of Cell Biology, School of Medicine, New York University, New York, NY, USA
| |
Collapse
|
48
|
Ning B, Zhao Y, Buza JA, Li W, Wang W, Jia T. Surgically‑induced mouse models in the study of bone regeneration: Current models and future directions (Review). Mol Med Rep 2017; 15:1017-1023. [PMID: 28138711 PMCID: PMC5367352 DOI: 10.3892/mmr.2017.6155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 12/13/2016] [Indexed: 01/17/2023] Open
Abstract
Bone regeneration has been extensively studied over the past several decades. The surgically‑induced mouse model is the key animal model for studying bone regeneration, of the various research strategies used. These mouse models mimic the trauma and recovery processes in vivo and serve as carriers for tissue engineering and gene modification to test various therapies or associated genes in bone regeneration. The present review introduces a classification of surgically induced mouse models in bone regeneration, evaluates the application and value of these models and discusses the potential development of further innovations in this field in the future.
Collapse
Affiliation(s)
- Bin Ning
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Yunpeng Zhao
- Department of Orthopedic Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - John A Buza
- Department of Orthopedic Surgery, New York University Medical Center, New York, NY 10003, USA
| | - Wei Li
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Wenzhao Wang
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Tanghong Jia
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
49
|
Yue R, Shen B, Morrison SJ. Clec11a/osteolectin is an osteogenic growth factor that promotes the maintenance of the adult skeleton. eLife 2016; 5. [PMID: 27976999 PMCID: PMC5158134 DOI: 10.7554/elife.18782] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/01/2016] [Indexed: 12/12/2022] Open
Abstract
Bone marrow stromal cells maintain the adult skeleton by forming osteoblasts throughout life that regenerate bone and repair fractures. We discovered that subsets of these stromal cells, osteoblasts, osteocytes, and hypertrophic chondrocytes secrete a C-type lectin domain protein, Clec11a, which promotes osteogenesis. Clec11a-deficient mice appeared developmentally normal and had normal hematopoiesis but reduced limb and vertebral bone. Clec11a-deficient mice exhibited accelerated bone loss during aging, reduced bone strength, and delayed fracture healing. Bone marrow stromal cells from Clec11a-deficient mice showed impaired osteogenic differentiation, but normal adipogenic and chondrogenic differentiation. Recombinant Clec11a promoted osteogenesis by stromal cells in culture and increased bone mass in osteoporotic mice in vivo. Recombinant human Clec11a promoted osteogenesis by human bone marrow stromal cells in culture and in vivo. Clec11a thus maintains the adult skeleton by promoting the differentiation of mesenchymal progenitors into mature osteoblasts. In light of this, we propose to call this factor Osteolectin. DOI:http://dx.doi.org/10.7554/eLife.18782.001
Collapse
Affiliation(s)
- Rui Yue
- Department of Pediatrics and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Bo Shen
- Department of Pediatrics and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Sean J Morrison
- Department of Pediatrics and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
50
|
Systemic and Local Administration of Antimicrobial and Cell Therapies to Prevent Methicillin-Resistant Staphylococcus epidermidis-Induced Femoral Nonunions in a Rat Model. Mediators Inflamm 2016; 2016:9595706. [PMID: 27478310 PMCID: PMC4961811 DOI: 10.1155/2016/9595706] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 12/27/2022] Open
Abstract
S. epidermidis is responsible for biofilm-related nonunions. This study compares the response to S. epidermidis-infected fractures in rats systemically or locally injected with vancomycin or bone marrow mesenchymal stem cells (BMSCs) in preventing the nonunion establishment. The 50% of rats receiving BMSCs intravenously (s-rBMSCs) died after treatment. A higher cytokine trend was measured in BMSCs locally injected rats (l-rBMSCs) at day 3 and in vancomycin systemically injected rats (l-VANC) at day 7 compared to the other groups. At day 14, the highest cytokine values were measured in l-VANC and in l-rBMSCs for IL-10. µCT showed a good bony bridging in s-VANC and excellent both in l-VANC and in l-rBMSCs. The bacterial growth was lower in s-VANC and l-VANC than in l-rBMSCs. Histology demonstrated the presence of new woven bone in s-VANC and a more mature bony bridging was found in l-VANC. The l-rBMSCs showed a poor bony bridging of fibrovascular tissue. Our results could suggest the synergic use of systemic and local injection of vancomycin as an effective treatment to prevent septic nonunions. This study cannot sustain the systemic injection of BMSCs due to high risks, while a deeper insight into local BMSCs immunomodulatory effects is mandatory before developing cell therapies in clinics.
Collapse
|