1
|
Huang S, Wen J, Zhang Y, Bai X, Cui ZK. Choosing the right animal model for osteomyelitis research: Considerations and challenges. J Orthop Translat 2023; 43:47-65. [PMID: 38094261 PMCID: PMC10716383 DOI: 10.1016/j.jot.2023.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 03/22/2024] Open
Abstract
Osteomyelitis is a debilitating bone disorder characterized by an inflammatory process involving the bone marrow, bone cortex, periosteum, and surrounding soft tissue, which can ultimately result in bone destruction. The etiology of osteomyelitis can be infectious, caused by various microorganisms, or noninfectious, such as chronic nonbacterial osteomyelitis (CNO) and chronic recurrent multifocal osteomyelitis (CRMO). Researchers have turned to animal models to study the pathophysiology of osteomyelitis. However, selecting an appropriate animal model that accurately recapitulates the human pathology of osteomyelitis while controlling for multiple variables that influence different clinical presentations remains a significant challenge. In this review, we present an overview of various animal models used in osteomyelitis research, including rodent, rabbit, avian/chicken, porcine, minipig, canine, sheep, and goat models. We discuss the characteristics of each animal model and the corresponding clinical scenarios that can provide a basic rationale for experimental selection. This review highlights the importance of selecting an appropriate animal model for osteomyelitis research to improve the accuracy of the results and facilitate the development of novel treatment and management strategies.
Collapse
Affiliation(s)
| | | | - Yiqing Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhong-Kai Cui
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
2
|
Vandenbulcke F, Anzillotti G, Ravasio G, Malagoli E, Conte P, Balzarini B, Kirienko A, Kon E. External fixator-assisted plating osteosynthesis in a rabbit model of femoral bone defects appears to be a feasible and reproducible surgical technique: preliminary insights from a bone substitute study. J Exp Orthop 2023; 10:77. [PMID: 37526773 PMCID: PMC10393914 DOI: 10.1186/s40634-023-00644-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 08/02/2023] Open
Abstract
PURPOSE The aims of this study were to assess the complications associated with the use of an external fixator-assisted plate osteosynthesis technique to stabilize a femoral bone defect in a rabbit model and to evaluate if this technique could avoid the mispositioning and the displacement of the femoral fragments during the surgical procedure. METHODS A preliminary cadaveric animal study was conducted to develop a new technique of external fixator-assisted plating. Thirty rabbits underwent a surgical procedure consisting in the creation of a femoral bone defect and, subsequently an implantation of a bone substitute through the assistance of a temporary external fixator. The fixator's ability to maintain length and alignment during surgery was documented. All intraoperative complications were prospectively collected. RESULTS No complications related to the use of the temporary external fixator were reported. The technique successfully prevented mispositioning and dislocation during plating in all the rabbits. CONCLUSION In a rabbit animal model, the use of an external fixator-assisted plate osteosynthesis technique appears to be feasible and effective in avoiding misposition and rotation of femoral fragments when performing osteotomy and plating to create a mid-diaphyseal femoral defect.
Collapse
Affiliation(s)
- F Vandenbulcke
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, 20089, Milan, Italy.
- Humanitas Clinical and Research Center, External Fixation Unit (Dr Kirienko A), Via Manzoni 56, Rozzano, 20089, Milan, Italy.
| | - G Anzillotti
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, 20089, Milan, Italy
- Humanitas Clinical and Research Center, External Fixation Unit (Dr Kirienko A), Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - G Ravasio
- Department of Veterinary Medicine, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
- University of Milan, Veterinary Teaching Hospital, Via Dell'Università 6, 26900, Lodi, Italy
| | - E Malagoli
- Humanitas Clinical and Research Center, External Fixation Unit (Dr Kirienko A), Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - P Conte
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, 20089, Milan, Italy
- Humanitas Clinical and Research Center, External Fixation Unit (Dr Kirienko A), Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - B Balzarini
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, 20089, Milan, Italy
- Humanitas Clinical and Research Center, External Fixation Unit (Dr Kirienko A), Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - A Kirienko
- Humanitas Clinical and Research Center, External Fixation Unit (Dr Kirienko A), Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - E Kon
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, 20089, Milan, Italy
- Humanitas Clinical and Research Center, External Fixation Unit (Dr Kirienko A), Via Manzoni 56, Rozzano, 20089, Milan, Italy
| |
Collapse
|
3
|
Zhao X, Li L, Chen M, Xu Y, Zhang S, Chen W, Liang W. Nanotechnology Assisted Targeted Drug Delivery for Bone Disorders: Potentials and Clinical Perspectives. Curr Top Med Chem 2020; 20:2801-2819. [PMID: 33076808 DOI: 10.2174/1568026620666201019110459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 04/26/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022]
Abstract
Nanotechnology and its allied modalities have brought revolution in tissue engineering and bone healing. The research on translating the findings of the basic and preclinical research into clinical practice is ongoing. Advances in the synthesis and design of nanomaterials along with advances in genomics and proteomics, and tissue engineering have opened a bright future for bone healing and orthopedic technology. Studies have shown promising outcomes in the design and fabrication of porous implant substrates that can be exploited as bone defect augmentation and drug-carrier devices. However, there are dozens of applications in orthopedic traumatology and bone healing for nanometer-sized entities, structures, surfaces, and devices with characteristic lengths ranging from tens 10s of nanometers to a few micrometers. Nanotechnology has made promising advances in the synthesis of scaffolds, delivery mechanisms, controlled modification of surface topography and composition, and biomicroelectromechanical systems. This study reviews the basic and translational sciences and clinical implications of the nanotechnology in tissue engineering and bone diseases. Recent advances in NPs assisted osteogenic agents, nanocomposites, and scaffolds for bone disorders are discussed.
Collapse
Affiliation(s)
- Xiaofeng Zhao
- Department of Orthopaedics, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), 568# Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| | - Laifeng Li
- Department of Traumatic Orthopedics, Affiliated Jinan Third Hospital of Jining Medical University, Jinan 250132, Shandong Province, China
| | - Meikai Chen
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, Zhejiang Province, China
| | - Yifan Xu
- Department of Orthopaedics, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), 568# Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| | - Songou Zhang
- Department of Orthopaedics, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), 568# Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| | - Wangzhen Chen
- Department of Orthopaedics, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), 568# Zhongxing North Road, Shaoxing 312000, Zhejiang Province, China
| | - Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, Zhejiang Province, China
| |
Collapse
|
4
|
Wong RM, Li TK, Li J, Ho WT, Chow SKH, Leung SS, Cheung WH, Ip M. A systematic review on current osteosynthesis-associated infection animal fracture models. J Orthop Translat 2020; 23:8-20. [PMID: 32440511 PMCID: PMC7231979 DOI: 10.1016/j.jot.2020.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
Objective Osteosynthesis-associated infection is a challenging complication post fracture fixation, burdening the patients and the orthopaedic surgeons alike. A clinically relevant animal model is critical in devising new therapeutic strategies. Our aim was to perform a systematic review to evaluate existing preclinical models and identify their applications in aspects of animal selection, bacterial induction, fracture fixation and complications. Methods A systematic literature research was conducted in PubMed and Embase up to February 2020. A total of 31 studies were included. Information on the animal, bacterial induction, fracture fixation, healing result and complications were extracted. Results Animals selected included murine (23), rabbit (6), ewe (1) and goat (1). Larger animals had enabled the use of human-sized implant, however small animals were more economical and easier in handling. Staphylococcus aureus (S. aureus) was the most frequently chosen bacteria for induction. Bacterial inoculation dose ranged from 102-8 CFU. Consistent and replicable infections were observed from 104 CFU in general. Methods of inoculation included injections of bacterial suspension (20), placement of foreign objects (8) and pretreatment of implants with established biofilm (3). Intramedullary implants (13), plates and screws (18) were used in most models. Radiological (29) and histological evaluations (24) in osseous healing were performed. Complications such as instability of fracture fixation (7), unexpected surgical death (5), sepsis (1) and persistent lameness (1) were encountered. Conclusion The most common animal model is the S. aureus infected open fracture internally fixated. Replicable infections were mainly from 104 CFU of bacteria. However, with the increase in antibiotic resistance, future directions should explore polymicrobial and antibiotic resistant strains, as these will no doubt play a major role in bone infection. Currently, there is also a lack of osteoporotic bone infection models and the pathophysiology is unexplored, which would be important with our aging population. The translational potential of this article This systematic review provides an updated overview and compares the currently available animal models of osteosynthesis-associated infections. A discussion on future research directions and suggestion of animal model settings were made, which is expected to advance the research in this field.
Collapse
Affiliation(s)
- Ronald M.Y. Wong
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Tsz-kiu Li
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Jie Li
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Wing-Tung Ho
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Simon K.-H. Chow
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | | | - Wing-Hoi Cheung
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
- Corresponding author. Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Margaret Ip
- Department of Microbiology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
5
|
Venkatesan JK, Rey-Rico A, Cucchiarini M. Current Trends in Viral Gene Therapy for Human Orthopaedic Regenerative Medicine. Tissue Eng Regen Med 2019; 16:345-355. [PMID: 31413939 PMCID: PMC6675832 DOI: 10.1007/s13770-019-00179-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/09/2019] [Accepted: 01/12/2019] [Indexed: 12/29/2022] Open
Abstract
Background Viral vector-based therapeutic gene therapy is a potent strategy to enhance the intrinsic reparative abilities of human orthopaedic tissues. However, clinical application of viral gene transfer remains hindered by detrimental responses in the host against such vectors (immunogenic responses, vector dissemination to nontarget locations). Combining viral gene therapy techniques with tissue engineering procedures may offer strong tools to improve the current systems for applications in vivo. Methods The goal of this work is to provide an overview of the most recent systems exploiting biomaterial technologies and therapeutic viral gene transfer in human orthopaedic regenerative medicine. Results Integration of tissue engineering platforms with viral gene vectors is an active area of research in orthopaedics as a means to overcome the obstacles precluding effective viral gene therapy. Conclusions In light of promising preclinical data that may rapidly expand in a close future, biomaterial-guided viral gene therapy has a strong potential for translation in the field of human orthopaedic regenerative medicine.
Collapse
Affiliation(s)
- Jagadeesh Kumar Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr, Bldg 37, 66421 Homburg/Saar, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr, Bldg 37, 66421 Homburg/Saar, Germany
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr, Bldg 37, 66421 Homburg/Saar, Germany
| |
Collapse
|
6
|
Off-Label Use of Bone Morphogenetic Protein 2 in the Reconstructions of Mandibular Continuity Defects. J Craniofac Surg 2017; 28:227-230. [PMID: 27977485 DOI: 10.1097/scs.0000000000003291] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
This paper describes 3 patients of off-label use of bone morphogenetic protein 2 (rhBMP-2) in the reconstruction of mandibular continuity defects. In the first patient, rhBMP-2 was associated with iliac crest bone graft for late mandibular reconstruction after resection of osteosarcoma. In the 2 other patients, rhBMP-2 was used alone. In 1 patient the mandibular continuity defect was due to resection for treatment of osteomyelitis and in the other patient a continuity defect was created by unsuccessful osteogenic distraction for correction of mandibular hypoplasia. Despite the good results in those patients, the off-label use of rhBMP-2 is associated with increased rate of complications, so more studies are needed to assess the predictability of the use of rhBMP-2 in mandibular continuity defects. Therefore, at the moment the off-label use of rhBMP-2 should be restricted to complicated bone defects in which the conventional alternatives of reconstruction were unsuccessful.
Collapse
|
7
|
Bara JJ, Dresing I, Zeiter S, Anton M, Daculsi G, Eglin D, Nehrbass D, Stadelmann VA, Betts DC, Müller R, Alini M, Stoddart MJ. A doxycycline inducible, adenoviral bone morphogenetic protein-2 gene delivery system to bone. J Tissue Eng Regen Med 2017; 12:e106-e118. [PMID: 27957814 DOI: 10.1002/term.2393] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/06/2016] [Accepted: 12/06/2016] [Indexed: 12/19/2022]
Abstract
We report the novel use of a tuneable, non-integrating viral gene delivery system to bone that can be combined with clinically approved biomaterials in an 'off-the-shelf' manner. Specifically, a doxycycline inducible Tet-on adenoviral vector (AdTetBMP-2) in combination with mesenchymal stromal cells (MSCs), fibrin and a biphasic calcium phosphate ceramic (MBCP®) was used to repair large bone defects in nude rats. Bone morphogenetic protein-2 (BMP-2) transgene expression could be effectively tuned by modification of the doxycycline concentration. The effect of adenoviral BMP-2 gene delivery upon bone healing was investigated in vivo in 4 mm critically sized, internally fixated, femoral defects. MSCs were transduced either by direct application of AdTetBMP-2 or by pre-coating MBCP granules with the virus. Radiological assessment scores post-mortem were significantly improved upon delivery of AdTetBMP-2. In AdTetBMP-2 groups, histological analysis revealed significantly more newly formed bone at the defect site compared with controls. Newly formed bone was vascularized and fully integrated with nascent tissue and implanted biomaterial. Improvement in healing outcome was achieved using both methods of vector delivery (direct application vs. pre-coating MCBP). Adenoviral delivery of BMP-2 enhanced bone regeneration achieved by the transplantation of MSCs, fibrin and MBCP in vivo. Importantly, our in vitro and in vivo data suggest that this can be achieved with relatively low (ng/ml) levels of the growth factor. Our model and novel gene delivery system may provide a powerful standardized tool for the optimization of growth factor delivery and release for the healing of large bone defects. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | - Iska Dresing
- AO Research Institute Davos, Davos Platz, Switzerland
| | | | - Martina Anton
- Klinikum Rechts der Isar der Technischen Universität München, Institute of Experimental Oncology and Therapy Research, Munich, Germany
| | - Guy Daculsi
- INSERM U791 Laboratory for Osteoarticular and Dental Tissue Engineering, Dental Faculty, Nantes University, Nantes, France
| | - David Eglin
- AO Research Institute Davos, Davos Platz, Switzerland
| | - Dirk Nehrbass
- AO Research Institute Davos, Davos Platz, Switzerland
| | | | - Duncan C Betts
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Davos Platz, Switzerland
| | | |
Collapse
|
8
|
|
9
|
Pilge H, Bittersohl B, Schneppendahl J, Hesper T, Zilkens C, Ruppert M, Krauspe R, Jäger M. Bone Marrow Aspirate Concentrate in Combination With Intravenous Iloprost Increases Bone Healing in Patients With Avascular Necrosis of the Femoral Head: A Matched Pair Analysis. Orthop Rev (Pavia) 2017; 8:6902. [PMID: 28507661 PMCID: PMC5402318 DOI: 10.4081/or.2016.6902] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/06/2016] [Indexed: 11/22/2022] Open
Abstract
With disease progression, avascular necrosis (AVN) of the femoral head may lead to a collapse of the articular surface. The exact pathophysiology of AVN remains unclear, although several conditions are known that can result in spontaneous cell death, leading to a reduction of trabecular bone and the development of AVN. Hip AVN treatment is stage-dependent in which two main stages of the disease can be distinguished: pre-collapse (ARCO 0-II) and post-collapse stage (ARCO III-IV, crescent sign). In the pre-collapse phase, core decompression (CD), with or without the addition of bone marrow (e.g. bone marrow aspirate concentrate, BMAC) or bone graft, is a common treatment alternative. In the post-collapse phase, THA (total hip arthroplasty) must be performed in most of the patients. In addition to surgical treatment, the intravenous application of Iloprost has been shown to have a curative potential and analgesic effect. From October 2009 to October 2014, 49 patients with AVN (stages I-III) were treated with core decompression at our institution. All patients were divided into group A (CD + BMAC) and group B (CD alone). Of these patients, 20 were included in a matched pair analysis. The patients were matched to age, gender, ARCO-stage, Kerboul combined necrotic angle, the cause of AVN, and whether Iloprost-therapy was performed. The Merle d’Aubigné Score and the Kerboul combined necrotic angle in a-p and lateral radiographs were evaluated pre- and postoperatively. The primary endpoint was a total hip arthroplasty. In group A, two patients needed THA while in group B four patients were treated with THA. In group A, the Merle d’Aubigné Score improved from 13.5 (pre-operatively) to 15.3 (postoperatively). In group B there was no difference between the pre- (14.3) and postoperative (14.1) assessment. The mean of the Kerboul angle showed no difference in both groups compared pre- to postoperatively (group A: pre-op 212°, postop 220°, group B: pre-op 213, postop 222°). Regarding radiographic evaluation, the interobserver variability revealed a moderate agreement between two raters regarding the pre-(ICC 0.594) and postoperative analysis (ICC 0.604).This study demonstrates that CD in combination with the application of autologous bone marrow aspirate concentrate into the femoral head seems to be a safe and efficient treatment alternative in the early stages of AVN of the femoral head when compared to CD alone.
Collapse
Affiliation(s)
- Hakan Pilge
- Department of Orthopedics, Düsseldorf University Clinic, Düsseldorf, Germany
| | - Bernd Bittersohl
- Department of Orthopedics, Düsseldorf University Clinic, Düsseldorf, Germany
| | | | - Tobias Hesper
- Department of Orthopedics, Düsseldorf University Clinic, Düsseldorf, Germany
| | - Christoph Zilkens
- Department of Orthopedics, Düsseldorf University Clinic, Düsseldorf, Germany
| | - Martin Ruppert
- Department of Orthopedics, Düsseldorf University Clinic, Düsseldorf, Germany
| | - Rüdiger Krauspe
- Department of Orthopedics, Düsseldorf University Clinic, Düsseldorf, Germany
| | - Marcus Jäger
- Department of Orthopedics and Traumatology, University Clinic Essen, Essen, Germany
| |
Collapse
|
10
|
Lovati AB, Romanò CL, Bottagisio M, Monti L, De Vecchi E, Previdi S, Accetta R, Drago L. Modeling Staphylococcus epidermidis-Induced Non-Unions: Subclinical and Clinical Evidence in Rats. PLoS One 2016; 11:e0147447. [PMID: 26796958 PMCID: PMC4721651 DOI: 10.1371/journal.pone.0147447] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 01/03/2016] [Indexed: 01/20/2023] Open
Abstract
S. epidermidis is one of the leading causes of orthopaedic infections associated with biofilm formation on implant devices. Open fractures are at risk of S. epidermidis transcutaneous contamination leading to higher non-union development compared to closed fractures. Although the role of infection in delaying fracture healing is well recognized, no in vivo models investigated the impact of subclinical low-grade infections on bone repair and non-union. We hypothesized that the non-union rate is directly related to the load of this commonly retrieved pathogen and that a low-grade contamination delays the fracture healing without clinically detectable infection. Rat femurs were osteotomized and stabilized with plates. Fractures were infected with a characterized clinical-derived methicillin-resistant S. epidermidis (103, 105, 108 colony forming units) and compared to uninfected controls. After 56 days, bone healing and osteomyelitis were clinically assessed and further evaluated by micro-CT, microbiological and histological analyses. The biofilm formation was visualized by scanning electron microscopy. The control group showed no signs of infection and a complete bone healing. The 103 group displayed variable response to infection with a 67% of altered bone healing and positive bacterial cultures, despite no clinical signs of infection present. The 105 and 108 groups showed severe signs of osteomyelitis and a non-union rate of 83–100%, respectively. The cortical bone reaction related to the periosteal elevation in the control group and the metal scattering detected by micro-CT represented limitations of this study. Our model showed that an intra-operative low-grade S. epidermidis contamination might prevent the bone healing, even in the absence of infectious signs. Our findings also pointed out a dose-dependent effect between the S. epidermidis inoculum and non-union rate. This pilot study identifies a relevant preclinical model to assess the role of subclinical infections in orthopaedic and trauma surgery and to test specifically designed diagnostic, prevention and therapeutic strategies.
Collapse
Affiliation(s)
- Arianna Barbara Lovati
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
- * E-mail:
| | - Carlo Luca Romanò
- Dipartimento di Chirurgia Ricostruttiva e delle Infezioni Osteo-articolari, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Marta Bottagisio
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
- Department of Veterinary Science and Public Health, University of Milan, Milan, Italy
| | - Lorenzo Monti
- Orthopaedics and Traumatology, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Elena De Vecchi
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Sara Previdi
- Laboratory of Cancer Cachexia AIRC Start-Up, Oncology Department, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Riccardo Accetta
- Orthopaedics and Traumatology, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Lorenzo Drago
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
- Department of Biomedical Science for Health, University of Milan, Milan, Italy
| |
Collapse
|
11
|
Repair of a Complicated Calvarial Defect: Reconstruction of an Infected Wound With rhBMP-2. Ann Plast Surg 2016; 76:205-10. [PMID: 26756598 DOI: 10.1097/sap.0000000000000515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Management of the previously infected craniofacial defect remains a significant clinical challenge, posing obstacles such as wound healing complications, lack of donor site availability, and predisposition to failure of the repair. Optimal therapy would reconstruct like with like, without donor site morbidity. The purpose of this study was to compare the efficacy of recombinant human bone morphogenetic protein-2 (rhBMP-2)-mediated bone regeneration with the current standard of autologous bone graft for repair of previously infected calvarial defects. METHODS Nineteen adult New Zealand white rabbits underwent subtotal calvariectomy. Bone flaps were inoculated with Staphylococcus aureus and replanted. After 1 week of infection, bone flaps were removed, and wounds were debrided, followed by 10 days of antibiotic treatment. After 6 weeks, animals underwent scar debridement followed by definitive reconstruction in 1 of 4 groups: empty control (n = 3), vehicle control (buffer solution on absorbable collagen sponge [ACS], n = 3), autologous bone graft (n = 3), or rhBMP-2 repair (rhBMP-2/ACS, n = 10). Animals underwent computed tomography imaging at 0, 2, 4, and 6 weeks postoperatively, followed by euthanization and histological analysis. Percent healing was determined by 3-dimensional analysis. A (time × group) 2-way analysis of variance was performed on healing versus treatment group and postoperative time. RESULTS At 6 weeks postoperatively, rhBMP-2/ACS and autologous bone graft resulted in 93% and 68% healing, respectively, whereas the empty and vehicle control treatment resulted in 27% and 26% healing (P < 0.001). Histologically, compared to autologous bone graft, bone in the rhBMP-2/ACS group was more cellular and more consistently continuous with wound margins. CONCLUSIONS The rhBMP-2 therapy is effective in achieving radiographic coverage of previously infected calvarial defects.
Collapse
|
12
|
Houdek MT, Wyles CC, Sierra RJ. Osteonecrosis of the femoral head: treatment with ancillary growth factors. Curr Rev Musculoskelet Med 2015; 8:233-9. [PMID: 25985987 PMCID: PMC4596200 DOI: 10.1007/s12178-015-9281-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Osteonecrosis (ON) of the femoral head, also known as avascular necrosis (AVN) of the femoral head, is a progressive disease that predominantly affects younger patients. During early stage of ON, decompression of the femoral head has been commonly used to improve pain. The decompression has been augmented with nonvascularized or vascularized bone grafts, mesenchymal stems cells, and growth factors. The use of adjuvant growth factors to supplement the core decompression has mainly been limited to animal models in an attempt to regenerate the necrotic lesion of ON. Factors utilized include bone morphogenetic proteins, vascular endothelial growth factors, hepatocyte growth factors, fibroblast growth factors, granulocyte colony-stimulating factors, and stem cells factors. In animal models, the use of these factors has been shown to increase bone formation and angiogenesis. Although promising, the use of these growth factors and cell-based therapies clinically remains limited.
Collapse
Affiliation(s)
- Matthew T. Houdek
- />Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905 USA
| | - Cody C. Wyles
- />Mayo Clinic Medical School, 200 First St. SW, Rochester, MN 55909 USA
| | - Rafael J. Sierra
- />Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905 USA
| |
Collapse
|
13
|
Scaffold-based anti-infection strategies in bone repair. Ann Biomed Eng 2014; 43:515-28. [PMID: 25476163 DOI: 10.1007/s10439-014-1205-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 11/26/2014] [Indexed: 12/14/2022]
Abstract
Bone fractures and non-union defects often require surgical intervention where biomaterials are used to correct the defect, and approximately 10% of these procedures are compromised by bacterial infection. Currently, treatment options are limited to sustained, high doses of antibiotics and surgical debridement of affected tissue, leaving a significant, unmet need for the development of therapies to combat device-associated biofilm and infections. Engineering implants to prevent infection is a desirable material characteristic. Tissue engineered scaffolds for bone repair provide a means to both regenerate bone and serve as a base for adding antimicrobial agents. Incorporating anti-infection properties into regenerative medicine therapies could improve clinical outcomes and reduce the morbidity and mortality associated with biomaterial implant-associated infections. This review focuses on current animal models and technologies available to assess bone repair in the context of infection, antimicrobial agents to fight infection, the current state of antimicrobial scaffolds, and future directions in the field.
Collapse
|
14
|
Abstract
Osteomyelitis of the foot and ankle is a common, potentially devastating condition with diagnostic and treatment challenges. Understanding the epidemiology and pathogenesis of osteomyelitis can raise clinical suspicion and guide testing and treatments. History and physical examination, laboratory studies, vascular studies, histologic and microbiologic analyses, and various imaging modalities contribute to diagnosis and treatment. Treatment including empiric broad-spectrum antibiotics and surgery should take a multidisciplinary approach to optimize patient factors, ensure eradication of the infection, and restore function. Optimization of vascular status, soft tissues, limb biomechanics, and physiologic state of the patient must be considered to accelerate and ensure healing.
Collapse
|
15
|
Evans C. Using genes to facilitate the endogenous repair and regeneration of orthopaedic tissues. INTERNATIONAL ORTHOPAEDICS 2014; 38:1761-9. [PMID: 25038968 DOI: 10.1007/s00264-014-2423-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 06/09/2014] [Indexed: 10/25/2022]
Abstract
Traditional tissue engineering approaches to the restoration of orthopaedic tissues promise to be expensive and not well suited to treating large numbers of patients. Advances in gene transfer technology offer the prospect of developing expedited techniques in which all manipulations can be performed percutaneously or in a single operation. This rests on the ability of gene delivery to provoke the sustained synthesis of relevant gene products in situ without further intervention. Regulated gene expression is also possible, but its urgency is reduced by our ignorance of exactly what levels and periods of expression are needed for specific gene products. This review describes various strategies by which gene therapy can be used to expedite the repair and regeneration of orthopaedic tissues. Strategies include the direct injection of vectors into sites of injury, the use of genetically modified, allogeneic cell lines and the intra-operative harvest of autologous tissues that are quickly transduced and returned to the body, either intact or following rapid cell isolation. Data obtained from pre-clinical experiments in animal models have provided much encouragement that such approaches may eventually find clinical application in human and veterinary medicine.
Collapse
Affiliation(s)
- Christopher Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA,
| |
Collapse
|
16
|
Reizner W, Hunter J, O’Malley N, Southgate R, Schwarz E, Kates S. A systematic review of animal models for Staphylococcus aureus osteomyelitis. Eur Cell Mater 2014; 27:196-212. [PMID: 24668594 PMCID: PMC4322679 DOI: 10.22203/ecm.v027a15] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Staphylococcus aureus (S. aureus) osteomyelitis is a significant complication for orthopaedic patients undergoing surgery, particularly with fracture fixation and arthroplasty. Given the difficulty in studying S. aureus infections in human subjects, animal models serve an integral role in exploring the pathogenesis of osteomyelitis, and aid in determining the efficacy of prophylactic and therapeutic treatments. Animal models should mimic the clinical scenarios seen in patients as closely as possible to permit the experimental results to be translated to the corresponding clinical care. To help understand existing animal models of S. aureus, we conducted a systematic search of PubMed and Ovid MEDLINE to identify in vivo animal experiments that have investigated the management of S. aureus osteomyelitis in the context of fractures and metallic implants. In this review, experimental studies are categorised by animal species and are further classified by the setting of the infection. Study methods are summarised and the relevant advantages and disadvantages of each species and model are discussed. While no ideal animal model exists, the understanding of a model's strengths and limitations should assist clinicians and researchers to appropriately select an animal model to translate the conclusions to the clinical setting.
Collapse
Affiliation(s)
| | | | | | | | | | - S.L. Kates
- Address for correspondence: Stephen L. Kates, 601 Elmwood Ave, Box 665, Rochester, NY 14642, USA,
| |
Collapse
|
17
|
Chen S, Yang J, Wang H, Chao Y, Zhang C, Shen J, Zhang P. Adenovirus encoding BMP-7 immobilized on titanium surface exhibits local delivery ability and regulates osteoblast differentiation in vitro. Arch Oral Biol 2013; 58:1225-31. [DOI: 10.1016/j.archoralbio.2013.03.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 03/20/2013] [Accepted: 03/31/2013] [Indexed: 10/26/2022]
|
18
|
Reifenrath J, Angrisani N, Lalk M, Besdo S. Replacement, refinement, and reduction: Necessity of standardization and computational models for long bone fracture repair in animals. J Biomed Mater Res A 2013; 102:2884-900. [DOI: 10.1002/jbm.a.34920] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/30/2013] [Accepted: 07/31/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Janin Reifenrath
- Small Animal Clinic; University of Veterinary Medicine Hannover; Bünteweg 9 30559 Hannover Germany
| | - Nina Angrisani
- Small Animal Clinic; University of Veterinary Medicine Hannover; Bünteweg 9 30559 Hannover Germany
| | - Mareike Lalk
- Small Animal Clinic; University of Veterinary Medicine Hannover; Bünteweg 9 30559 Hannover Germany
| | - Silke Besdo
- Institute of Continuum Mechanics; Leibniz Universität Hannover; Appelstr. 11 30167 Hannover Germany
| |
Collapse
|
19
|
Sun S, Guo H, Zhang J, Yu B, Sun K, Jin Q. Adenovirus-mediated expression of bone morphogenetic protein-2 activates titanium particle-induced osteoclastogenesis and this effect occurs in spite of the suppression of TNF-α expression by siRNA. Int J Mol Med 2013; 32:403-9. [PMID: 23708523 DOI: 10.3892/ijmm.2013.1392] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/13/2013] [Indexed: 11/05/2022] Open
Abstract
The phagocytosis of wear particles by macrophages results in the secretion of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), which play a major role in promoting osteoclast recruitment. The inhibition of TNF-α expression decreases osteoclastogenesis. In a previous study, we demonstrated that bone morphogenetic protein-2 (BMP-2) can activate wear debris-induced osteoclast recruitment in the presence of receptor activator of nuclear factor (NF)-κB ligand (RANKL); however, whether these effects are associated with pro-inflammatory cytokines remains unclear. In this study, we constructed an adenoviral vector carrying TNF-small interfering RNA (siRNA) (Ad-TNF-siRNA), as well as a vector carrying both the BMP-2 gene and TNF-α-siRNA (Ad-BMP-2-TNF-siRNA). The two adenoviral vectors significantly suppressed the expression of TNF-α; however, only treatment with Ad-TNF-siRNA significantly inhibited osteoclastogenesis. We demonstrate that the overexpression of BMP-2, despite the suppression of TNF-α expression by Ad-BMP-2-TNF-siRNA, increases the size and number of titanium (Ti) particle-induced multinuclear osteoclasts, the expression of osteoclast genes, as well as the resorption area. There were no differences observed between Ti particle-induced and Ad-BMP-2-TNF-siRNA-induced osteoclast formation. Moreover, Ad-BMP-2-TNF-siRNA directly acted upon osteoclast precursors by increasing the level of c-Fos, regulating other signaling pathways, such as p38 phosphorylated c-Jun N-terminal kinase (p-JNK) and phosphorylated IκB (p‑IκB). Taken together, these data demonstrate that treatment with Ad-BMP-2-TNF-siRNA increases wear debris-induced osteoclast formation by activating c-Fos and that these effects are not associated with pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Shouxuan Sun
- Department of Orthopedics, Affiliated Hospital of Ningxia Medical University, Ningxia 750004, P.R. China
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
Since the discovery of bone marrow derived stromal cell osteogenesis in the 1960s, tissue engineering with adult multipotent stromal cells (MSCs) has evolved as a promising approach to restore structure and function of bone compromised by injury or disease. To date, accelerated bone formation with MSCs has been demonstrated with a variety of tissue engineering strategies. Though MSC bone tissue engineering has advanced over the last few decades, limitations to clinical translation remain. A current review of this promising field is presented with a specific focus on equine investigations.
Collapse
Affiliation(s)
- Mandi J Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Veterinary Clinical Sciences Department, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| | | |
Collapse
|
21
|
Abstract
Gene delivery to bone is useful both as an experimental tool and as a potential therapeutic strategy. Among its advantages over protein delivery are the potential for directed, sustained and regulated expression of authentically processed, nascent proteins. Although no clinical trials have been initiated, there is a substantial pre-clinical literature documenting the successful transfer of genes to bone, and their intraosseous expression. Recombinant vectors derived from adenovirus, retrovirus and lentivirus, as well as non-viral vectors, have been used for this purpose. Both ex vivo and in vivo strategies, including gene-activated matrices, have been explored. Ex vivo delivery has often employed mesenchymal stem cells (MSCs), partly because of their ability to differentiate into osteoblasts. MSCs also have the potential to home to bone after systemic administration, which could serve as a useful way to deliver transgenes in a disseminated fashion for the treatment of diseases affecting the whole skeleton, such as osteoporosis or osteogenesis imperfecta. Local delivery of osteogenic transgenes, particularly those encoding bone morphogenetic proteins, has shown great promise in a number of applications where it is necessary to regenerate bone. These include healing large segmental defects in long bones and the cranium, as well as spinal fusion and treating avascular necrosis.
Collapse
Affiliation(s)
- C H Evans
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
22
|
King WJ, Krebsbach PH. Growth factor delivery: how surface interactions modulate release in vitro and in vivo. Adv Drug Deliv Rev 2012; 64:1239-56. [PMID: 22433783 PMCID: PMC3586795 DOI: 10.1016/j.addr.2012.03.004] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 02/24/2012] [Accepted: 03/05/2012] [Indexed: 02/06/2023]
Abstract
Biomaterial scaffolds have been extensively used to deliver growth factors to induce new bone formation. The pharmacokinetics of growth factor delivery has been a critical regulator of their clinical success. This review will focus on the surface interactions that control the non-covalent incorporation of growth factors into scaffolds and the mechanisms that control growth factor release from clinically relevant biomaterials. We will focus on the delivery of recombinant human bone morphogenetic protein-2 from materials currently used in the clinical practice, but also suggest how general mechanisms that control growth factor incorporation and release delineated with this growth factor could extend to other systems. A better understanding of the changing mechanisms that control growth factor release during the different stages of preclinical development could instruct the development of future scaffolds for currently untreatable injuries and diseases.
Collapse
Affiliation(s)
- William J. King
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, 1011 N. University Ave., Ann Arbor, MI 48109, USA
| | - Paul H. Krebsbach
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, 1011 N. University Ave., Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, 2200 Bonisteel, Blvd., Ann Arbor, MI 48109, USA
| |
Collapse
|
23
|
Abstract
This review is aimed at clinicians appraising preclinical trauma studies and researchers investigating compromised bone healing or novel treatments for fractures. It categorises the clinical scenarios of poor healing of fractures and attempts to match them with the appropriate animal models in the literature. We performed an extensive literature search of animal models of long bone fracture repair/nonunion and grouped the resulting studies according to the clinical scenario they were attempting to reflect; we then scrutinised them for their reliability and accuracy in reproducing that clinical scenario. Models for normal fracture repair (primary and secondary), delayed union, nonunion (atrophic and hypertrophic), segmental defects and fractures at risk of impaired healing were identified. Their accuracy in reflecting the clinical scenario ranged greatly and the reliability of reproducing the scenario ranged from 100% to 40%. It is vital to know the limitations and success of each model when considering its application.
Collapse
Affiliation(s)
- L. A. Mills
- Royal National Orthopaedic Hospital, Stanmore, Brockley
Hill, Middlesex HA7 4LP, UK
| | - A. H. R. W. Simpson
- Edinburgh University, Department
of Orthopaedics and Trauma, Chancellors Building, Little
France, Edinburgh EH16 4SB, UK
| |
Collapse
|
24
|
Yun YR, Jang JH, Jeon E, Kang W, Lee S, Won JE, Kim HW, Wall I. Administration of growth factors for bone regeneration. Regen Med 2012; 7:369-85. [DOI: 10.2217/rme.12.1] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Growth factors (GFs) such as BMPs, FGFs, VEGFs and IGFs have significant impacts on osteoblast behavior, and thus have been widely utilized for bone tissue regeneration. Recently, securing biological stability for a sustainable and controllable release to the target tissue has been a challenge to practical applications. This challenge has been addressed to some degree with the development of appropriate carrier materials and delivery systems. This review highlights the importance and roles of those GFs, as well as their proper administration for targeting bone regeneration. Additionally, the in vitro and in vivo performance of those GFs with or without the use of carrier systems in the repair and regeneration of bone tissue is systematically addressed. Moreover, some recent advances in the utility of the GFs, such as using fusion technology, are also reviewed.
Collapse
Affiliation(s)
- Ye-Rang Yun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 330-714, Korea
| | - Jun Hyeog Jang
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Eunyi Jeon
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Wonmo Kang
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Sujin Lee
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Jong-Eun Won
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 330-714, Korea
- Department of Nanobiomedical Science & WCU Research Center, Dankook University Graduate School, Cheonan 330-714, Korea
| | - Hae Won Kim
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 330-714, Korea
| | - Ivan Wall
- Department of Nanobiomedical Science & WCU Research Center, Dankook University Graduate School, Cheonan 330-714, Korea
- Department of Biochemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| |
Collapse
|
25
|
Ishihara A, Bertone AL. Cell-mediated and direct gene therapy for bone regeneration. Expert Opin Biol Ther 2012; 12:411-23. [PMID: 22324829 DOI: 10.1517/14712598.2012.661709] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Bone regeneration is required for the treatment of fracture non/delayed-unions and bone defects. However, most current treatment modalities have limited efficacy, and newer therapeutic strategies, such as gene therapy, have substantial benefit for bone repair and regeneration. AREAS COVERED This review discusses experimental and clinical applications of cell-mediated and direct gene therapy for bone regeneration. The review covers literature on this subject from 2000 to February 2012. EXPERT OPINION Direct gene therapy using various viral and non-viral vectors of cell-mediated genes has been demonstrated to induce bone regeneration, although use of such vectors has shown some risk in human application. Osteoinductive capability of a number of progenitor cells isolated from bone marrow, fat, muscle and skin tissues, has been demonstrated by genetic modification with osteogenic genes. Cell-mediated gene therapy using such osteogenic gene-expressing progenitor cells has shown promising results in promoting bone regeneration in extensive animal work in recent years.
Collapse
Affiliation(s)
- Akikazu Ishihara
- The Ohio State University, Department of Veterinary Clinical Sciences, Comparative Orthopedic Research Laboratories, Columbus, OH 43210, USA
| | | |
Collapse
|
26
|
Abstract
The current management for chronic osteomyelitis centers on adequate antibiotic coverage and surgical debridement of nonviable tissue. The eradication of osteomyelitis, however, often involves a prolonged and frustrating course of management. Nonsurgical adjunctive modalities have not been widely used, mostly due to a lack of perceived efficacy, and have remained in a state of infancy. In this article, we will outline the rationale, current status, and evidence for several potential adjuncts to osteomyelitis management.
Collapse
Affiliation(s)
- Robert C Fang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | |
Collapse
|
27
|
Long J, Li P, Du HM, Liu L, Zheng XH, Lin YF, Wang H, Jing W, Tang W, Chen WH, Tian WD. Effects of bone morphogenetic protein 2 gene therapy on new bone formation during mandibular distraction osteogenesis at rapid rate in rabbits. ORAL SURGERY, ORAL MEDICINE, ORAL PATHOLOGY, ORAL RADIOLOGY, AND ENDODONTICS 2011; 112:50-57. [PMID: 21194991 DOI: 10.1016/j.tripleo.2010.09.065] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 08/08/2010] [Accepted: 09/22/2010] [Indexed: 01/12/2023]
Abstract
OBJECTIVE We investigated the effect of recombinant human bone morphogenetic protein 2 (rhBMP-2) on new bone formation during rapid-rate mandibular distraction osteogenesis. We also explored the feasibility of using local BMP-2 gene therapy to compensate for bad callus formation caused by a rapid distraction rate. STUDY DESIGN Bone marrow mesenchymal stem cells (MSCs) from Japanese rabbits were transfected with adenovirus (adv)-BMP-2. The right mandibles of the rabbits were distracted after corticotomy. The distraction rate in group A was 0.8 mm/d. The distraction rate in group B was 2.4 mm/d, and the distraction gap was injected with adv-lacZ-transfected bone marrow MSCs. The distraction rate in group C was 2.4 mm/d, and the distraction gap was injected with adv-BMP-2-transfected bone marrow MSCs. New generation bone tissue in the distraction gap was analyzed by plain radiograph examinations, microfocus computerized tomography (micro-CT) examinations, and biomechanical tests at weeks 2, 4, and 8 of the consolidation period. RESULTS Radiographic and micro-CT examinations showed a better bone quality in group C compared with group A at weeks 2 and 4 of the consolidation period. There was no obvious new bone formation in group B. The trabecular parameters (trabecular thickness, trabecular number, volumetric bone mineral density at tissue, and bone volume fraction) were significantly higher in group C than in group A at weeks 2 and 4. At week 8, no significant difference were detected for all parameters except trabecular number between groups A and C. All biomechanical stress parameters were significantly higher in group C than in group A at week 4, and only peak stress was significantly different at week 8. CONCLUSIONS Gene therapy using rhBMP-2-modified MSCs promoted new bone formation during mandibular distraction osteogenesis, and effectively compensated for the detrimental effect of rapid distraction rate on new bone formation.
Collapse
Affiliation(s)
- Jie Long
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Weil YA, Rivkin G, Safran O, Liebergall M, Foldes AJ. The Outcome of Surgically Treated Femur Fractures Associated With Long-Term Bisphosphonate Use. ACTA ACUST UNITED AC 2011; 71:186-90. [DOI: 10.1097/ta.0b013e31821957e3] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Bae JH, Song HR, Kim HJ, Lim HC, Park JH, Liu Y, Teoh SH. Discontinuous release of bone morphogenetic protein-2 loaded within interconnected pores of honeycomb-like polycaprolactone scaffold promotes bone healing in a large bone defect of rabbit ulna. Tissue Eng Part A 2011; 17:2389-97. [PMID: 21682591 DOI: 10.1089/ten.tea.2011.0032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The choice of an appropriate carrier and its microarchitectural design is integral in directing bone ingrowth into the defect site and determining its subsequent rate of bone formation and remodeling. We have selected a three-dimensional polycaprolactone (PCL) scaffold with an interconnected honeycomb-like porous structure to provide a conduit for vasculature ingrowth as well as an osteoconductive pathway to guide recruited cells responding to a unique triphasic release of osteoinductive bone morphogenetic proteins (BMP) from these PCL scaffolds. We hypothesize that the use of recombinant human bone morphogenetic protein 2 (rhBMP2)-PCL constructs promotes rapid union and bone regeneration of a large defect. Results of our pilot study on a unilateral 15 mm mid-diaphyseal segmental rabbit ulna defect demonstrated enhanced bone healing with greater amount of bone formation and bridging under plain radiography and microcomputed tomography imaging when compared with an empty PCL and untreated group after 8 weeks postimplantation. Quantitative measurements showed significantly higher bone volume fraction and trabecular thickness, with lower trabecular separation in the rhBMP2-treated groups. Histology evaluation also revealed greater mature bone formation spanning across the entire scaffold region compared with other groups, which showed no bone regeneration within the central defect zone. We highlight that it is the uniqueness of the scaffold having a highly porous network of channels that promoted vascular integration and allowed for cellular infiltration, leading to a discontinuous triphasic BMP2 release profile that mimicked the release profile during natural repair mechanisms in vivo. This study serves as preclinical evidence demonstrating the potential of combining osteoinductive rhBMP2 with our PCL constructs for the repair of large defects in a large animal model.
Collapse
Affiliation(s)
- Ji-Hoon Bae
- Department of Orthopaedic Surgery, Korea University Ansan Hospital, Ansan Si, Gyeonggi Do, Korea
| | | | | | | | | | | | | |
Collapse
|
30
|
Kuo HC, Chiu CC, Chang WC, Sheen JM, Ou CY, Kuo HC, Chen RF, Hsu TY, Chang JC, Hsaio CC, Wang FS, Huang CC, Huang HY, Yang KD. Use of Proteomic Differential Displays to Assess Functional Discrepancies and Adjustments of Human Bone Marrow- and Wharton Jelly-Derived Mesenchymal Stem Cells. J Proteome Res 2011; 10:1305-15. [DOI: 10.1021/pr101057w] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hsing-Chun Kuo
- Institute of Nursing and Department of Nursing, Chang Gung Institute of Technology Chia-Yi Campus, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abdelmagid SM, Barbe MF, Hadjiargyrou M, Owen TA, Razmpour R, Rehman S, Popoff SN, Safadi FF. Temporal and spatial expression of osteoactivin during fracture repair. J Cell Biochem 2011; 111:295-309. [PMID: 20506259 DOI: 10.1002/jcb.22702] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We previously identified osteoactivin (OA) as a novel secreted osteogenic factor with high expression in developing long bones and calvaria, and that stimulates osteoblast differentiation and matrix mineralization in vitro. In this study, we report on OA mRNA and protein expression in intact long bone and growth plate, and in fracture calluses collected at several time points up to 21 days post-fracture (PF). OA mRNA and protein were highly expressed in osteoblasts localized in the metaphysis of intact tibia, and in hypertrophic chondrocytes localized in growth plate, findings assessed by in situ hybridization and immunohistochemistry, respectively. Using a rat fracture model, Northern blot analysis showed that expression of OA mRNA was significantly higher in day-3 and day-10 PF calluses than in intact rat femurs. Using in situ hybridization, we examined OA mRNA expression during fracture healing and found that OA was temporally regulated, with positive signals seen as early as day-3 PF, reaching a maximal intensity at day-10 PF, and finally declining at day-21 PF. At day-5 PF, which correlates with chondrogenesis, OA mRNA levels were significantly higher in the soft callus than in intact femurs. Similarly, we detected high OA protein immunoexpression throughout the reparative phase of the hard callus compared to intact femurs. Interestingly, the secreted OA protein was also detected within the newly made cartilage matrix and osteoid tissue. Taken together, these results suggest the possibility that OA plays an important role in bone formation and serves as a positive regulator of fracture healing.
Collapse
Affiliation(s)
- Samir M Abdelmagid
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Self-complementary AAV2.5-BMP2-coated femoral allografts mediated superior bone healing versus live autografts in mice with equivalent biomechanics to unfractured femur. Mol Ther 2011; 19:1416-25. [PMID: 21206485 DOI: 10.1038/mt.2010.294] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Structural allografts used for critical bone defects have limited osteogenic properties for biointegration. Although ex vivo tissue-engineered constructs expressing bone morphogenetic protein-2 (BMP2) have demonstrated efficacy in critical defect models, similar success has not been achieved with off-the-shelf acellular approaches, including allografts coated with freeze-dried single-stranded adeno-associated virus (ssAAV-BMP2). To see whether the self-complementary AAV serotype 2.5 vector (scAAV2.5-BMP2) could overcome this, we performed side-by-side comparisons in vitro and in the murine femoral allograft model. Although ssAAV-BMP2 was unable to induce BMP2 expression and differentiation of C3H10T1/2 cells in culture, scAAV2.5-BMP2 transduction led to dose-dependent BMP2 expression and alkaline phosphatase activity, and displayed a 25-fold increased transduction efficiency in vivo. After 6 weeks, the ssAAV-BMP2 coating failed to demonstrate any significant effects. However, all allografts coated with 10(10) scAAV2.5-BMP2 formed a new cortical shell that was indistinguishable to that formed by live autografts. Additionally, coated allografts experienced reduced resorption resulting in a threefold increase in graft bone volume versus autograft. This led to biomechanical superiority versus both allografts and autografts, and equivalent torsional rigidity to unfractured femur. Collectively, these results demonstrate that scAAV2.5-BMP2 coating overcomes the major limitations of structural allografts, which can be used to heal critical defects of any size.
Collapse
|
33
|
Dong JL, Li LX, Mu WD, Wang YH, Zhou DS, Wei Hao, Zou DB, Kun Hu, Ji Li, Cui FZ. Bone Regeneration with BMP-2 Gene-modified Mesenchymal Stem Cells Seeded on Nano-hydroxyapatite/Collagen/ Poly(L-Lactic Acid) Scaffolds. J BIOACT COMPAT POL 2010. [DOI: 10.1177/0883911510380436] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this study, the capacity of bone morphogenetic protein 2 (BMP-2) gene-transfected bone marrow-derived mesenchymal stem cells (MSCs) in combination with nano-hydroxyapatite/collagen/poly(L-lactic acid) (nHAC/ PLA) to improve the repair of bone defects in rabbit was explored. MSCs from New Zealand White rabbits were cultured and injected with pIRES2-EGFPhBMP-2 or pIRES2-EGFP by electroporation. After the transfer efficiency was determined through the expression of EGFP, the MSCs were seeded on scaffolds to generate an in vitro 3D cell/scaffold construct. The adhesion and proliferation of the MSCs cultured in the scaffold was assessed by SEM. The cellular constructs obtained were allografted into the 15 mm critical-sized segmental bone defects in the radius of New Zealand White rabbits for 12 weeks. The bone regeneration was assessed by radiographical and histological analyses. In vitro, nHAC/PLA facilitated MSC adhesion and proliferation on the scaffold, and gene transfer efficiency reached a maximum of 35.5 ± 3.8%. In vivo, the implantation of BMP-2 transfected MSCs/nHAC/PLA construct significantly enhanced the formation of new bone in the segmental defect, compared to the control groups. This novel 3D BMP-2 transfected MSCs/nHAC/PLA construct has the potential for bone repair by genetic tissue engineering approach.
Collapse
Affiliation(s)
- Jin-Lei Dong
- Department of Orthopedics, Provincial Hospital Affiliated to Shandong University, Jinan 250021, People's Republic of China
| | - Lian-Xin Li
- Department of Orthopedics, Provincial Hospital Affiliated to Shandong University, Jinan 250021, People's Republic of China
| | - Wei-Dong Mu
- Department of Orthopedics, Provincial Hospital Affiliated to Shandong University, Jinan 250021, People's Republic of China
| | - Yong-Hui Wang
- Department of Orthopedics, Provincial Hospital Affiliated to Shandong University, Jinan 250021, People's Republic of China
| | - Dong-Sheng Zhou
- Department of Orthopedics, Provincial Hospital Affiliated to Shandong University, Jinan 250021, People's Republic of China,
| | - Wei Hao
- Department of Spinal Cord Injury, General Hospital of Ji'nan Military Area, Jinan 250031, People's Republic of China
| | - De-Bo Zou
- Department of Orthopedics, Qianfoshan Hospital Affiliated to Shandong University, Jinan 250014, People's Republic of China
| | - Kun Hu
- Institute of Nuclear and New Energy Technology Tsinghua University, Beijing 100084, People's Republic of China
| | - Ji Li
- Department of Orthopedics, Jinan Fourth Hospital Jinan 250031, People's Republic of China
| | - Fu-Zhai Cui
- State Key Laboratory of New Ceramics and Fine Processing Department of Materials Science and Engineering, Tsinghua University Beijing 100084, People's Republic of China,
| |
Collapse
|
34
|
Abstract
Clinical problems in bone healing include large segmental defects, spinal fusions, and the nonunion and delayed union of fractures. Gene-transfer technologies have the potential to aid healing by permitting the local delivery and sustained expression of osteogenic gene products within osseous lesions. Key questions for such an approach include the choice of transgene, vector and gene-transfer strategy. Most experimental data have been obtained using cDNAs encoding osteogenic growth factors such as bone morphogenetic protein-2 (BMP-2), BMP-4 and BMP-7, in conjunction with both nonviral and viral vectors using in vivo and ex vivo delivery strategies. Proof of principle has been convincingly demonstrated in small-animal models. Relatively few studies have used large animals, but the results so far are encouraging. Once a reliable method has been developed, it will be necessary to perform detailed pharmacological and toxicological studies, as well as satisfy other demands of the regulatory bodies, before human clinical trials can be initiated. Such studies are very expensive and often protracted. Thus, progress in developing a clinically useful gene therapy for bone healing is determined not only by scientific considerations, but also by financial constraints and the ambient regulatory environment.
Collapse
|
35
|
Pelled G, Ben-Arav A, Hock C, Reynolds DG, Yazici C, Zilberman Y, Gazit Z, Awad H, Gazit D, Schwarz EM. Direct gene therapy for bone regeneration: gene delivery, animal models, and outcome measures. TISSUE ENGINEERING PART B-REVIEWS 2010; 16:13-20. [PMID: 20143927 DOI: 10.1089/ten.teb.2009.0156] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
While various problems with bone healing remain, the greatest clinical change is the absence of an effective approach to manage large segmental defects in limbs and craniofacial bones caused by trauma or cancer. Thus, nontraditional forms of medicine, such as gene therapy, have been investigated as a potential solution. The use of osteogenic genes has shown great potential in bone regeneration and fracture healing. Several methods for gene delivery to the fracture site have been described. The majority of them include a cellular component as the carrying vector, an approach known as cell-mediated gene therapy. Yet, the complexity involved with cell isolation and culture emphasizes the advantages of direct gene delivery as an alternative strategy. Here we review the various approaches of direct gene delivery for bone repair, the choice of animal models, and the various outcome measures required to evaluate the efficiency and safety of each technique. Special emphasis is given to noninvasive, quantitative, in vivo monitoring of gene expression and biodistribution in live animals. Research efforts should aim at inducing a transient, localized osteogenic gene expression within a fracture site to generate an effective therapeutic approach that would eventually lead to clinical use.
Collapse
Affiliation(s)
- Gadi Pelled
- Skeletal Biotechnology Laboratory, Hebrew University of Jerusalem-Hadassah Medical Campus, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Abstract
Nanotechnology and its attendant techniques have yet to make a significant impact on the science of bone healing. However, the potential benefits are immediately obvious with the result that hundreds of researchers and firms are performing the basic research needed to mature this nascent, yet soon to be fruitful niche. Together with genomics and proteomics, and combined with tissue engineering, this is the new face of orthopaedic technology. The concepts that orthopaedic surgeons recognize are fabrication processes that have resulted in porous implant substrates as bone defect augmentation and medication-carrier devices. However, there are dozens of applications in orthopaedic traumatology and bone healing for nanometer-sized entities, structures, surfaces, and devices with characteristic lengths ranging from 10s of nanometers to a few micrometers. Examples include scaffolds, delivery mechanisms, controlled modification of surface topography and composition, and biomicroelectromechanical systems. We review the basic science, clinical implications, and early applications of the nanotechnology revolution and emphasize the rich possibilities that exist at the crossover region between micro- and nanotechnology for developing new treatments for bone healing.
Collapse
|
38
|
Abstract
Alteration of the bone healing process with bone morphogenetic proteins offers a new perspective in orthopaedic surgery in those adverse situations that necessitate bone grafting. BMPs have been demonstrated to be effective and safe for human application and have an efficacy comparable with that of autologous bone grafting. Nevertheless, clinical trials with level 1 evidence are still limited in their ability to extrapolate robust and safe clinical conclusions for the possible indications mentioned in this article. Future research should refine issues regarding the relative effectiveness of bone morphogenetic proteins, the interaction between bone morphogenetic protein subtypes, and their specific effect on various target cell populations.
Collapse
|
39
|
Abstract
The management of open fractures continues to be complicated by high rates of treatment failure and significant patient disability and dissatisfaction. The use of bone morphogenetic proteins (BMPs) in the treatment of open fractures has been assessed by a number of different clinical trials, both in the acute management of open fractures and in the delayed reconstruction of bone defects secondary to open fracture. This review describes the scientific basis for the use of BMPs in open fractures, reviews the current evidence for their use in open fractures, provides grades of recommendation for the different uses of BMPs in open fractures, and identifies important areas for additional research.
Collapse
|
40
|
Calori GM, Donati D, Di Bella C, Tagliabue L. Bone morphogenetic proteins and tissue engineering: future directions. Injury 2009; 40 Suppl 3:S67-76. [PMID: 20082795 DOI: 10.1016/s0020-1383(09)70015-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
As long as bone repair and regeneration is considered as a complex clinical condition, the administration of more than one factor involved in fracture healing might be necessary. The effectiveness or not of bone morphogenetic proteins (BMPs) in association with other growth factors and with mesenchymal stem cells in bone regeneration for fracture healing and bone allograft integration is of great interest to the scientific community. In this study we point out possible future developments in BMPs, concerning research and clinical applications.
Collapse
Affiliation(s)
- G M Calori
- Orthopaedic Institute Gaetano Pini, University of Milan, Italy.
| | | | | | | |
Collapse
|
41
|
Ability of recombinant human bone morphogenetic protein 2 to enhance bone healing in the presence of tobramycin: evaluation in a rat segmental defect model. J Orthop Trauma 2009; 23:693-701. [PMID: 19858977 DOI: 10.1097/bot.0b013e3181b01b2f] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine whether locally applied tobramycin influences the ability of recombinant human bone morphogenetic protein 2 (rhBMP-2) to heal a segmental defect in the rat femur. METHODS The influence of tobramycin on the osteogenic differentiation of mesenchymal stem cells was first evaluated in vitro. For the subsequent, in vivo experiments, a 5-mm segmental defect was created in the right femur of each of 25 Sprague-Dawley rats and stabilized with an external fixator and four Kirschner wires. Rats were divided in four groups: empty control, tobramycin (11 mg)/absorbable collagen sponge, rhBMP-2 (11 microg)/absorbable collagen sponge, and rhBMP-2/absorbable collagen sponge with tobramycin. Bone healing was monitored by radiography at 3 and 8 weeks. Animals were euthanized at 8 weeks and the properties of the defect were compared with the intact contralateral femur. Bone formation in the defect region was assessed by dual-energy x-ray absorptiometry, microcomputed tomography, histology, and mechanical testing. RESULTS Tobramycin exerted a dose-dependent inhibition of alkaline phosphatase induction and calcium deposition by mesenchymal stem cells cultured under osteogenic conditions. The inhibition was reversed in the presence of 500 ng/mL of rhBMP-2. Segmental defects in the rat femora failed to heal in the absence of rhBMP-2. Tobramycin exerted no inhibitory effects on the ability of rhBMP-2 to heal these defects and increased the bone area of the defects treated with rhBMP-2. Data obtained from all other parameters of healing, including dual-energy x-ray absorptiometry, microcomputed tomography, histology, and mechanical testing, were unaffected by tobramycin. CONCLUSIONS Although our in vitro results suggested that tobramycin inhibits the osteogenic differentiation of mesenchymal stem cells, this could be overcome by rhBMP-2. Tobramycin did not impair the ability of rhBMP-2 to heal critical-sized femoral defects in rats. Indeed, bone area was increased by nearly 20% in the rhBMP-2 group treated with tobramycin. This study shows that locally applied tobramycin can be used in conjunction with rhBMP-2 to enhance bone formation at fracture sites.
Collapse
|
42
|
Pelled G, Ben-Arav A, Hock C, Reynolds DG, Yazici C, Zilberman Y, Gazit Z, Awad H, Gazit D, Schwarz EM. Direct Gene Therapy for Bone Regeneration: Gene Delivery, Animal Models, and Outcome Measures. Tissue Eng Part A 2009. [DOI: 10.1089/ten.tea.2009.0156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
43
|
Autografts and Xenografts of Skin Fibroblasts Delivering BMP-2 Effectively Promote Orthotopic and Ectopic Osteogenesis. Anat Rec (Hoboken) 2009; 292:777-86. [DOI: 10.1002/ar.20904] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
44
|
Li BC, Zhang JJ, Xu C, Zhang LC, Kang JY, Zhao H. Treatment of rabbit femoral defect by firearm with BMP-4 gene combined with TGF-beta1. ACTA ACUST UNITED AC 2009; 66:450-6. [PMID: 19204520 DOI: 10.1097/ta.0b013e3181848cd6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Firearm bone fractures are difficult to treat compared with general ones as both soft tissue and bone are injured more extensively and severely with contamination in the wound track. The bone morphogenetic protein (BMP) and transforming growth factor (TGF)-beta play an important role in bone fracture healing. Therefore, BMP-4 combined with TGF-beta1 was used to improve and accelerate the repair of rabbit femoral defect resulting from firearm. METHODS Femoral defect was made with 0.375 g steel ball fired at 350 m/s. At 6 hours after wounding, the debridement and irrigation were performed, followed by trimming the ends of defected bone at day 7. Plasmid-encoded BMP-4 gene identified in vitro and TGF-beta1 were injected into the tissue of upper and lower parts and the epicenter of the defected area at 2 weeks after wounding, again TGF-beta1 was given at 5 weeks. At 3, 7, 11, and 15 weeks after wounding, the expression of mRNA and protein of BMP-4 were detected by reverse transcription-polymerase chain reaction and Western blot. The activity of alkaline phosphatase and calcium content were measured for describing osteogenetic ability. The course and quality of osteogenesis were determined quantitatively by pathohistological and X-ray examinations. RESULTS In vivo BMP-4 mRNA and protein could be continually expressed for 8 weeks. The determination of alkaline phosphatase activity and calcium content showed osteogenetic ability was significantly enhanced by BMP-4 gene combined with TGF-beta1. The pathohistological and X-ray examinations revealed that osteogenetic speed was prominently accelerated, and the quality was improved after the treatment. CONCLUSION The repair of rabbit femoral defect resulting from firearm can be significantly improved and accelerated by BMP-4 gene combined with TGF-beta1.
Collapse
Affiliation(s)
- Bing Cang Li
- Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China.
| | | | | | | | | | | |
Collapse
|
45
|
Li X, Wang J, Ni Y, Yan X, Lu Q, Xu H, Cheng Q, Liu K. Bone morphogenetic protein-2 stimulation of cartilage regeneration in canine tracheal graft. J Heart Lung Transplant 2009; 28:285-9. [PMID: 19285622 DOI: 10.1016/j.healun.2008.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 07/16/2008] [Accepted: 12/01/2008] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Graft stenosis is among the most serious post-surgical complications that can occur after tracheal transplantation. Typically, stenosis is caused by resorption of tracheal cartilage. Bone morphogenetic protein-2 (BMP-2) is efficient at stimulating bone or cartilage regeneration. In this study, BMP-2 is tested for its effects on stimulation of cartilage regeneration in tracheal transplantation. METHODS For tracheal autotransplantation, 24 mongrel dogs were divided equally into four groups and BMP-2 was injected between the cartilage rings at doses of 1, 3, 5 or 7 mg. For tracheal allotransplantation, 12 mongrel dogs were divided equally into two groups. One group received 5 mg of BMP-2 per graft, and the other received collagen only as a control. The grafts were harvested after 4 weeks and subjected to pathologic analysis. The diameter of the graft lumen and areas of new cartilage regeneration were measured. RESULTS Regenerated cartilage areas were found in both the injected area and around the perichondrium. The areas of regenerated cartilage, as well as the diameter of the tracheal lumen, increased significantly with increasing concentrations of BMP-2. Five milligrams per milliliter was the most effective dose of BMP-2 in this study. CONCLUSIONS BMP-2 can significantly stimulate cartilage regeneration in tracheal grafts and also can be used to prevent stenosis after tracheal transplantation.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Evans CH, Ghivizzani SC, Robbins PD. Orthopedic gene therapy in 2008. Mol Ther 2009; 17:231-44. [PMID: 19066598 PMCID: PMC2835052 DOI: 10.1038/mt.2008.265] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Accepted: 10/26/2008] [Indexed: 02/07/2023] Open
Abstract
Orthopedic disorders, although rarely fatal, are the leading cause of morbidity and impose a huge socioeconomic burden. Their prevalence will increase dramatically as populations age and gain weight. Many orthopedic conditions are difficult to treat by conventional means; however, they are good candidates for gene therapy. Clinical trials have already been initiated for arthritis and the aseptic loosening of prosthetic joints, and the development of bone-healing applications is at an advanced, preclinical stage. Other potential uses include the treatment of Mendelian diseases and orthopedic tumors, as well as the repair and regeneration of cartilage, ligaments, and tendons. Many of these goals should be achievable with existing technologies. The main barriers to clinical application are funding and regulatory issues, which in turn reflect major safety concerns and the opinion, in some quarters, that gene therapy should not be applied to nonlethal, nongenetic diseases. For some indications, advances in nongenetic treatments have also diminished enthusiasm. Nevertheless, the preclinical and early clinical data are impressive and provide considerable optimism that gene therapy will provide straightforward, effective solutions to the clinical management of several common debilitating disorders that are otherwise difficult and expensive to treat.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Molecular Orthopaedics, Harvard Medical School, Boston, Massachusetts, USA.
| | | | | |
Collapse
|
47
|
Kanellakopoulou K, Galanopoulos I, Soranoglou V, Tsaganos T, Tziortzioti V, Maris I, Papalois A, Giamarellou H, Giamarellos-Bourboulis EJ. Treatment of experimental osteomyelitis caused by methicillin-resistant Staphylococcus aureus with a synthetic carrier of calcium sulphate (Stimulan) releasing moxifloxacin. Int J Antimicrob Agents 2008; 33:354-9. [PMID: 19097865 DOI: 10.1016/j.ijantimicag.2008.09.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 09/12/2008] [Accepted: 09/12/2008] [Indexed: 11/25/2022]
Abstract
The objectives of this study were to assess the efficacy of a synthetic semihydrate form of calcium sulphate (Stimulan) in experimental bone infection caused by methicillin-resistant Staphylococcus aureus (MRSA). Osteomyelitis was induced after inoculation of the test pathogen in the left tibia of 72 New Zealand rabbits assigned to the following groups: 18 control rabbits (Group A); 18 rabbits with Stimulan implanted (Group B); and 36 rabbits with moxifloxacin-impregnated Stimulan implanted (Group C). Rabbits were sacrificed at weekly intervals and cancellous bone was harvested for histopathology and for estimation of bacterial growth and concentrations of moxifloxacin. Bacterial growth from cancellous bone of Group C was significantly lower than the respective growth of Groups A and B on all days of sacrifice. The main histological finding of animals in all three groups was a moderate to intense inflammatory reaction accompanied by fibrosis. The degree of fibrosis was higher in Group C compared with both other groups. Infiltration by giant cells was also observed, which was greater in Group C on Day 42. Antibiotic levels in bone were higher for bone samples closer to the site of implantation. In conclusion, Stimulan admixed with 10% moxifloxacin was very effective in achieving complete eradication of the causative pathogen in experimental osteomyelitis caused by MRSA.
Collapse
|
48
|
Tsialogiannis E, Polyzois I, Tang QO, Pavlou G, Tsiridis E, Heliotis M, Tsiridis E. Targeting bone morphogenetic protein antagonists:in vitroandin vivoevidence of their role in bone metabolism. Expert Opin Ther Targets 2008; 13:123-37. [DOI: 10.1517/14728220802637725] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
49
|
Santoni BG, Simon Turner A, Wheeler DL, Nicholas RW, Anchordoquy TJ, Ehrhart N. Gene therapy to enhance allograft incorporation after host tissue irradiation. Clin Orthop Relat Res 2008; 466:1921-9. [PMID: 18506562 PMCID: PMC2584270 DOI: 10.1007/s11999-008-0297-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Accepted: 04/28/2008] [Indexed: 01/31/2023]
Abstract
Structural bone allografts are used to reconstruct large skeletal defects after tumor surgery. Although allograft-related complications are declining, the use of perioperative radiation therapy is associated with a poorer outcome. Recently, BMP-2 levels in the host bed were reportedly diminished after exposure to radiation doses consistent with those used perioperatively to treat musculoskeletal sarcoma. Reintroduction of this osteogenic protein may circumvent the deleterious effects of preoperative radiation on allograft incorporation. We introduced a novel polymeric BMP-2 gene delivery system into the host-allograft junctions at the time of transplantation in an ovine tibial defect model with or without preoperative exposure to 50 Gy radiation. After 4 months, we noted no radiographic or histologic improvements in allograft incorporation after preoperative radiation and BMP-2 reintroduction; however, 50 Gy radiation was associated with increased porosity in the interface regions and poorer radiographic healing. We identified no BMP2-expressing cells or protein in the interface at the study end point, suggesting the polymeric gene delivery system was unable to promote extended expression of the protein or induce a healing response. Although gene therapy may hold promise as a novel technique to improve allograft incorporation, our data do not support that contention with the current approach.
Collapse
Affiliation(s)
- Brandon G. Santoni
- Department of Clinical Sciences, Colorado State University Veterinary Medical Center, Fort Collins, CO USA
| | - A. Simon Turner
- Department of Clinical Sciences, Colorado State University Veterinary Medical Center, Fort Collins, CO USA
| | | | | | - Tom J. Anchordoquy
- Department of Pharmaceutical Sciences, University of Colorado Health Sciences Center, Denver, CO USA
| | - Nicole Ehrhart
- Department of Clinical Sciences, Colorado State University Veterinary Medical Center, Fort Collins, CO USA ,Department of Clinical Sciences, James L. Voss Veterinary Medical Center, Colorado State University, Fort Collins, CO 80523 USA
| |
Collapse
|
50
|
Stevens B, Yang Y, Mohandas A, Stucker B, Nguyen KT. A review of materials, fabrication methods, and strategies used to enhance bone regeneration in engineered bone tissues. J Biomed Mater Res B Appl Biomater 2008; 85:573-82. [PMID: 17937408 DOI: 10.1002/jbm.b.30962] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Over the last decade, bone engineered tissues have been developed as alternatives to autografts and allografts to repair and reconstruct bone defects. This article provides a review of the current technologies in bone tissue engineering. Factors used for fabrication of three-dimensional bone scaffolds such as materials, cells, and biomolecular signals, as well as required properties for ideal bone scaffolds, are reviewed. In addition, current fabrication techniques including rapid prototyping are elaborated upon. Finally, this review article further discusses some effective strategies to enhance cell ingrowth in bone engineered tissues; for example, nanotopography, biomimetic materials, embedded growth factors, mineralization, and bioreactors. In doing so, it suggests that there is a possibility to develop bone substitutes that can repair bone defects and promote new bone formation for orthopedic applications.
Collapse
Affiliation(s)
- Brian Stevens
- Department of Biological and Irrigation Engineering, Utah State University, Logan, Utah, USA
| | | | | | | | | |
Collapse
|