1
|
Gao Q, Jiang Y, Zhou D, Li G, Han Y, Yang J, Xu K, Jing Y, Bai L, Geng Z, Zhang H, Zhou G, Zhu M, Ji N, Han R, Zhang Y, Li Z, Wang C, Hu Y, Shen H, Wang G, Shi Z, Han Q, Chen X, Su J. Advanced glycation end products mediate biomineralization disorder in diabetic bone disease. Cell Rep Med 2024; 5:101694. [PMID: 39173634 PMCID: PMC11524989 DOI: 10.1016/j.xcrm.2024.101694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 06/04/2024] [Accepted: 07/26/2024] [Indexed: 08/24/2024]
Abstract
Patients with diabetes often experience fragile fractures despite normal or higher bone mineral density (BMD), a phenomenon termed the diabetic bone paradox (DBP). The pathogenesis and therapeutics opinions for diabetic bone disease (DBD) are not fully explored. In this study, we utilize two preclinical diabetic models, the leptin receptor-deficient db/db mice (DB) mouse model and the streptozotocin-induced diabetes (STZ) mouse model. These models demonstrate higher BMD and lower mechanical strength, mirroring clinical observations in diabetic patients. Advanced glycation end products (AGEs) accumulate in diabetic bones, causing higher non-enzymatic crosslinking within collagen fibrils. This inhibits intrafibrillar mineralization and leads to disordered mineral deposition on collagen fibrils, ultimately reducing bone strength. Guanidines, inhibiting AGE formation, significantly improve the microstructure and biomechanical strength of diabetic bone and enhance bone fracture healing. Therefore, targeting AGEs may offer a strategy to regulate bone mineralization and microstructure, potentially preventing the onset of DBD.
Collapse
Affiliation(s)
- Qianmin Gao
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Yingying Jiang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China.
| | - Dongyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Yafei Han
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Jingzhi Yang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China; Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Guangyin Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Mengru Zhu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Ning Ji
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Ruina Han
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Yuanwei Zhang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Zuhao Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Chuandong Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Hao Shen
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Zhongmin Shi
- Department of Orthopedics, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Qinglin Han
- Orthopaedic Department, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China.
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China; Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China.
| |
Collapse
|
2
|
Glatt V, Bartnikowski N, Bartnikowski M, Aguilar L, Schuetz M, Tetsworth K. Intramedullary implant stability affects patterns of fracture healing in mice with morphologically different bone phenotypes. Bone 2024; 179:116978. [PMID: 37993038 DOI: 10.1016/j.bone.2023.116978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/18/2023] [Accepted: 11/19/2023] [Indexed: 11/24/2023]
Abstract
Almost all prior mouse fracture healing models have used needles or K-wires for fixation, unwittingly providing inadequate mechanical stability during the healing process. Our contention is that the reported outcomes have predominantly reflected this instability, rather than the impact of diverse biological conditions, pharmacologic interventions, exogenous growth factors, or genetic considerations. This important issue becomes obvious upon a critical review of the literature. Therefore, the primary aim of this study was to demonstrate the significance of mouse-specific implants designed to provide both axial and torsional stability (Screw and IM Nail) compared to conventional pins (Needle and K-wires), even when used in mice with differently sized marrow canals and diverse genetic backgrounds. B6 (large medullary canal), DBA, and C3H (smaller medullary canals) mice were employed, all of which have different bone morphologies. Closed femoral fractures were created and stabilized with intramedullary implants that provide different mechanical conditions during the healing process. The most important finding of this study was that appropriately designed mouse-specific implants, providing both axial and torsional stability, had the greatest influence on bone healing outcomes regardless of the different bone morphologies encountered. For instance, unstable implants in the B6 strain (largest medullary canal) resulted in significantly greater callus, with a fracture region mainly comprising trabecular bone along with the presence of cartilage 28 days after surgery. The DBA and C3H strains (with smaller medullary canals) instead formed significantly less callus, and only had a small amount of intracortical trabeculation remaining. Moreover, with more stable fracture fixation a higher BV/TV was observed and cortices were largely restored to their original dimensions and structure, indicating an accelerated healing and remodeling process. These observations reveal that the diaphyseal cortical thickness, influenced by the genetic background of each strain, played a pivotal role in determining the amount of bone formation in response to the fracture. These findings are highly important, indicating the rate and type of tissue formed is a direct result of mechanical instability, and this most likely would mask the true contribution of the tested genes, genetic backgrounds, or various therapeutic agents administered during the bone healing process.
Collapse
Affiliation(s)
- Vaida Glatt
- Department of Orthopaedic Surgery, University of Texas Health Science Center, San Antonio, TX, United States of America; Sam and Ann Barshop Institute for Longevity and Aging Studies at the University of Texas Health Science Center, San Antonio, TX, United States of America; Queensland University of Technology, Brisbane, Australia; Orthopaedic Research Centre of Australia, Brisbane, Australia.
| | | | | | - Leonardo Aguilar
- Department of Orthopaedic Surgery, University of Texas Health Science Center, San Antonio, TX, United States of America; Sam and Ann Barshop Institute for Longevity and Aging Studies at the University of Texas Health Science Center, San Antonio, TX, United States of America
| | | | - Kevin Tetsworth
- Department of Orthopaedic Surgery, The Royal Brisbane and Women's Hospital, Brisbane, Australia; Orthopaedic Research Centre of Australia, Brisbane, Australia
| |
Collapse
|
3
|
Menger MM, Stief M, Scheuer C, Rollmann MF, Herath SC, Braun BJ, Ehnert S, Nussler AK, Menger MD, Laschke MW, Histing T. Diclofenac, a NSAID, delays fracture healing in aged mice. Exp Gerontol 2023; 178:112201. [PMID: 37169100 DOI: 10.1016/j.exger.2023.112201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs), such as diclofenac, belong to the most prescribed analgesic medication after traumatic injuries. However, there is accumulating evidence that NSAIDs impair fracture healing. Because bone regeneration in aged patients is subject to significant changes in cell differentiation and proliferation as well as a markedly altered pharmacological action of drugs, we herein analyzed the effects of diclofenac on bone healing in aged mice using a stable closed femoral facture model. Thirty-three mice (male n = 14, female n = 19) received a daily intraperitoneal injection of diclofenac (5 mg/kg body weight). Vehicle-treated mice (n = 29; male n = 13, female n = 16) served as controls. Fractured mice femora were analyzed by means of X-ray, biomechanics, micro computed tomography (μCT), histology and Western blotting. Biomechanical analyses revealed a significantly reduced bending stiffness in diclofenac-treated animals at 5 weeks after fracture when compared to vehicle-treated controls. Moreover, the callus tissue in diclofenac-treated aged animals exhibited a significantly reduced amount of bone tissue and higher amounts of fibrous tissue. Further histological analyses demonstrated less lamellar bone after diclofenac treatment, indicating a delay in callus remodeling. This was associated with a decreased number of osteoclasts and an increased expression of osteoprotegerin (OPG) during the early phase of fracture healing. These findings indicate that diclofenac delays fracture healing in aged mice by affecting osteogenic growth factor expression and bone formation as well as osteoclast activity and callus remodeling.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany; Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany.
| | - Maximilian Stief
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Claudia Scheuer
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Mika F Rollmann
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany
| | - Steven C Herath
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany
| | - Benedikt J Braun
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany; Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Andreas K Nussler
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany; Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
4
|
Constant C, Moriarty TF, Arens D, Pugliese B, Zeiter S. Peri-anesthetic hypothermia in rodents: A factor to consider for accurate and reproducible outcomes in orthopedic device-related infection studies. J Orthop Res 2023; 41:619-628. [PMID: 35716157 DOI: 10.1002/jor.25397] [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: 01/19/2022] [Revised: 05/12/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023]
Abstract
Orthopedic device-related infection (ODRI) preclinical models are widely used in translational research. Most ODRI models require induction of general anesthesia, which frequently results in hypothermia in rodents. This study aimed to evaluate the impact of peri-anesthetic hypothermia in rodents on outcomes in preclinical ODRI studies. A retrospective analysis of all rodents that underwent surgery under general anesthesia to induce an ODRI model with inoculation of Staphylococcus epidermidis between 2016 and 2020 was conducted. A one-way multivariate analysis of covariance (one-way MANCOVA) was used to determine the fixed effect of peri-anesthetic hypothermia (hypothermic defined as rectal temperature <35°C) on the combined harvested tissue and implant colony-forming unit (CFU) counts, and having controlled for the study groups including treatments received, duration of surgery and anesthesia, and study period. The results showed a significant effect of peri-anesthetic hypothermia on the post-mortem combined CFU counts from the harvested tissue and implant(s) (p = 0.01) when comparing normo- versus hypothermic rodents. Using Wilks' Λ as a criterion to determine the contribution of independent variables to the model, peri-anesthetic hypothermia was the most significant, though still a weak predictor, of increased harvested CFU counts. Altogether, the data corroborate the concept that bacterial colonization is affected by abnormal body temperature during general anesthesia at the time of bacterial inoculation in rodents, which needs to be taken into consideration to decrease infection data variability and improve experimental reproducibility.
Collapse
|
5
|
He C, Lv Q, Liu Z, Long S, Li H, Xiao Y, Yang X, Liu Y, Liu C, Wang Z. Random and aligned electrostatically spun PLLA nanofibrous membranes enhance bone repair in mouse femur midshaft defects. J Biomater Appl 2023; 37:1582-1592. [PMID: 36662630 DOI: 10.1177/08853282221144220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Long-segment bone defects are a common clinical challenge and abstract biomaterials are a promising therapy. Poly-L-lactic acid (PLLA) nanofibrous membranes prepared by electrostatic spinning have a good bone repair potential. However, there are random and aligned surface morphologies of electrostatic spun PLLA nanofibrous membranes, which can affect the migration, proliferation, and differentiation ability of cells. The role of surface morphology in the repair of long bone defects in vivo is currently unknown. In this study, random and aligned electrostatically spun PLLA nanofibrous membranes were prepared, characterised, and implanted into a femur midshaft defect mouse model. The ability of electrostatically spun PLLA nanofibrous membranes to enhance bone repair was tested using X-ray photography, high-resolution micro-computed tomography (micro-CT), and pathological section specimens. The results showed that both random and aligned electrostatically spun PLLA nanofibrous membranes enhanced bone regeneration at bone defects, but the aligned ones exhibited superior results. These results provide a theoretical basis for engineering the surface morphology of bone repair materials.
Collapse
Affiliation(s)
- Chengkai He
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Basic Medical School of Kunming Medical University, Kunming, China
| | - Qiong Lv
- Outpatient Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhui Liu
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shengyu Long
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haohan Li
- The First Clinical College of Kunming Medical University, Kunming, China
| | - Ya Xiao
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Xingyu Yang
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Yuhang Liu
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Cai Liu
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Zhihua Wang
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,Yunnan Provincial Clinical Medical for Bone and Joint Diseases, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
6
|
Menger MM, Körbel C, Bauer D, Bleimehl M, Tobias AL, Braun BJ, Herath SC, Rollmann MF, Laschke MW, Menger MD, Histing T. Photoacoustic imaging for the study of oxygen saturation and total hemoglobin in bone healing and non-union formation. PHOTOACOUSTICS 2022; 28:100409. [PMID: 36213763 PMCID: PMC9535319 DOI: 10.1016/j.pacs.2022.100409] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 08/14/2022] [Accepted: 09/25/2022] [Indexed: 06/16/2023]
Abstract
Non-union formation represents a major complication in trauma surgery. Adequate vascularization has been recognized as vital for bone healing. However, the role of vascularization in the pathophysiology of non-union formation remains elusive. This is due to difficulties in studying bone microcirculation in vivo. Therefore, we herein studied in a murine osteotomy model whether photoacoustic imaging may be used to analyze vascularization in bone healing and non-union formation. We found that oxygen saturation within the callus tissue is significantly lower in non-unions compared to unions and further declines over time. Moreover, the amount of total hemoglobin (HbT) within the callus tissue was markedly reduced in non-unions. Correlation analyses showed a strong positive correlation between microvessel density and HbT, indicating that photoacoustically determined HbT is a valid parameter to assess vascularization during bone healing. In summary, photoacoustic imaging is a promising approach to study vascular function and tissue oxygenation in bone regeneration.
Collapse
Affiliation(s)
- Maximilian M. Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Christina Körbel
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - David Bauer
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Michelle Bleimehl
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Anne L. Tobias
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Benedikt J. Braun
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
| | - Steven C. Herath
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
| | - Mika F. Rollmann
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
| | - Matthias W. Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Michael D. Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
7
|
Schmitz N, Timmen M, Kostka K, Hoerr V, Schwarz C, Faber C, Hansen U, Matthys R, Raschke MJ, Stange R. A novel MRI compatible mouse fracture model to characterize and monitor bone regeneration and tissue composition. Sci Rep 2020; 10:16238. [PMID: 33004928 PMCID: PMC7529903 DOI: 10.1038/s41598-020-73301-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
Over the last years, murine in vivo magnetic resonance imaging (MRI) contributed to a new understanding of tissue composition, regeneration and diseases. Due to artefacts generated by the currently used metal implants, MRI is limited in fracture healing research so far. In this study, we investigated a novel MRI-compatible, ceramic intramedullary fracture implant during bone regeneration in mice. Three-point-bending revealed a higher stiffness of the ceramic material compared to the metal implants. Electron microscopy displayed a rough surface of the ceramic implant that was comparable to standard metal devices and allowed cell attachment and growth of osteoblastic cells. MicroCT-imaging illustrated the development of the callus around the fracture site indicating a regular progressing healing process when using the novel implant. In MRI, different callus tissues and the implant could clearly be distinguished from each other without any artefacts. Monitoring fracture healing using MRI-compatible implants will improve our knowledge of callus tissue regeneration by 3D insights longitudinal in the same living organism, which might also help to reduce the consumption of animals for future fracture healing studies, significantly. Finally, this study may be translated into clinical application to improve our knowledge about human bone regeneration.
Collapse
Affiliation(s)
- Nina Schmitz
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Münster, Germany
| | - Melanie Timmen
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine, University Muenster, Albert-Schweitzer-Campus 1, W1, 48149, Münster, Germany
| | - Katharina Kostka
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine, University Muenster, Albert-Schweitzer-Campus 1, W1, 48149, Münster, Germany
| | - Verena Hoerr
- Translational Research Imaging Center, Clinic of Radiology, University Hospital Muenster, Münster, Germany
| | - Christian Schwarz
- Translational Research Imaging Center, Clinic of Radiology, University Hospital Muenster, Münster, Germany
| | - Cornelius Faber
- Translational Research Imaging Center, Clinic of Radiology, University Hospital Muenster, Münster, Germany
| | - Uwe Hansen
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine, University Muenster, Albert-Schweitzer-Campus 1, W1, 48149, Münster, Germany
| | | | - Michael J Raschke
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Münster, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine, University Muenster, Albert-Schweitzer-Campus 1, W1, 48149, Münster, Germany.
| |
Collapse
|
8
|
Gunderson ZJ, Campbell ZR, McKinley TO, Natoli RM, Kacena MA. A comprehensive review of mouse diaphyseal femur fracture models. Injury 2020; 51:1439-1447. [PMID: 32362447 PMCID: PMC7323889 DOI: 10.1016/j.injury.2020.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 04/08/2020] [Indexed: 02/07/2023]
Abstract
Complications related to treatment of long bone fractures still stand as a major challenge for orthopaedic surgeons. Elucidation of the mechanisms of bone healing and development, and the subsequent alteration of these mechanisms to improve outcomes, typically requires animal models as an intermediary between in vitro and human clinical studies. Murine models are some of the most commonly used in translational research, and mouse fracture models are particularly diverse, offering a wide variety of customization with distinct benefits and limitations depending on the study. This review critically examines three common femur fracture models in the mouse, namely cortical hole, 3-point fracture (Einhorn), and segmental bone defect. We lay out the general procedure for execution of each model, evaluate the practical implications and important advantages/disadvantages of each and describe recent innovations. Furthermore, we explore the applications that each model is best adapted for in the context of the current state of murine orthopaedic research.
Collapse
Affiliation(s)
- Zachary J. Gunderson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Zachery R. Campbell
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Todd O. McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Roman M. Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Melissa A. Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA,Richard L. Roudebush VA Medical Center, IN, USA,Corresponding Author: Melissa A. Kacena, Ph.D., Director of Basic and Translational Research, Professor of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN 46202, (317) 278-3482 – office, (317) 278-9568 – fax
| |
Collapse
|
9
|
Collier CD, Hausman BS, Zulqadar SH, Din ES, Anderson JM, Akkus O, Greenfield EM. Characterization of a reproducible model of fracture healing in mice using an open femoral osteotomy. Bone Rep 2020; 12:100250. [PMID: 32090156 PMCID: PMC7025178 DOI: 10.1016/j.bonr.2020.100250] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/26/2020] [Accepted: 02/03/2020] [Indexed: 01/03/2023] Open
Abstract
Purpose The classic fracture model, described by Bonnarens and Einhorn in 1984, enlists a blunt guillotine to generate a closed fracture in a pre-stabilized rodent femur. However, in less experienced hands, this technique yields considerable variability in fracture pattern and requires highly-specialized equipment. This study describes a reproducible and low-cost model of mouse fracture healing using an open femoral osteotomy. Methods Femur fractures were produced in skeletally mature male and female mice using an open femoral osteotomy after intramedullary stabilization. Mice were recovered for up to 28 days prior to analysis with microradiographs, histomorphometry, a novel μCT methodology, and biomechanical torsion testing at weekly intervals. Results Eight mice were excluded due to complications (8/193, 4.1%), including unacceptable fracture pattern (2/193, 1.0%). Microradiographs showed progression of the fracture site to mineralized callus by 14 days and remodelling 28 days after surgery. Histomorphometry from 14 to 28 days revealed decreased cartilage area and maintained bone area. μCT analysis demonstrated a reduction in mineral surface from 14 to 28 days, stable mineral volume, decreased strut number, and increased strut thickness. Torsion testing at 21 days showed that fractured femurs had 61% of the ultimate torque, 63% of the stiffness, and similar twist to failure when compared to unfractured contralateral femurs. Conclusions The fracture model described herein, an open femoral osteotomy, demonstrated healing comparable to that reported using closed techniques. This simple model could be used in future research with improved reliability and reduced costs compared to the current options. This study characterized a simple and reproducible model of fracture healing in mice using an open femoral osteotomy. Analysis by x-ray, histomorphometry, µCT, and biomechanical testing demonstrated healing comparable to current models. This simple model could be used to increase investigation into fracture healing, delayed union, and non-union.
Collapse
Affiliation(s)
- C D Collier
- Department of Orthopaedics, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - B S Hausman
- Department of Orthopaedics, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - S H Zulqadar
- Department of Orthopaedics, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - E S Din
- Department of Orthopaedics, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - J M Anderson
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - O Akkus
- Department of Orthopaedics, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - E M Greenfield
- Department of Orthopaedics, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
10
|
Profiling microRNA expression in murine bone healing and non-union formation: Role of miR-140 during the early stage of bone healing. PLoS One 2019; 14:e0218395. [PMID: 31323027 PMCID: PMC6641081 DOI: 10.1371/journal.pone.0218395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/31/2019] [Indexed: 11/19/2022] Open
Abstract
Although cellular and molecular mechanisms during the course of bone healing have been thoroughly investigated, the regulation of gene expression by microRNA during bone regeneration is still poorly understood. We hypothesized that nonunion formation is associated with different microRNA expression patterns and that target proteins of these microRNAs are differently expressed in callus tissue of nonunions compared to physiologically healing bones. In a well-established femoral osteotomy model in CD-1 mice osteotomies were induced which result either in healing or in nonunion formation. MicroRNA and target protein expression was evaluated by microarray, quantitative real-time polymerase chain reaction (qrt-PCR) and Western blot. Microarray analyses demonstrated 44 microRNAs to be relevant for nonunion formation compared to physiological bone healing. In nonunions qrt-PCR could validate a higher expression of microRNA-140-3p and microRNA-140-5p. This was associated with a reduced expression of Dnpep and stromal cell-derived factor (SDF)-1α, which are both known to be target proteins of microRNA-140 and also to be involved in the process of bone healing. These data suggest that an increased expression of microRNA-140-3p and microRNA-140-5p markedly contributes to the development of nonunions, most probably by affecting bone morphogenetic protein (BMP)-2 function during the early stage of healing due to a reduced SDF-1α expression.
Collapse
|
11
|
Development of a novel murine delayed secondary fracture healing in vivo model using periosteal cauterization. Arch Orthop Trauma Surg 2019; 139:1743-1753. [PMID: 31399754 PMCID: PMC6825648 DOI: 10.1007/s00402-019-03255-y] [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: 11/29/2018] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Delayed union and nonunion development remain a major clinical problematic complication during fracture healing, with partially unclear pathophysiology. Incidences range from 5 to 40% in high-risk patients, such as patients with periosteal damage. The periosteum is essential in adequate fracture healing, especially during soft callus formation. In this study, we hypothesize that inducing periosteal damage in a murine bone healing model will result in a novel delayed union model. MATERIALS AND METHODS A mid-shaft femoral non-critically sized osteotomy was created in skeletally mature C57BL/6 mice and stabilized with a bridging plate. In half of the mice, a thin band of periosteum adjacent to the osteotomy was cauterized. Over 42 days of healing, radiographic, biomechanical, micro-computed tomography and histological analysis was performed to assess the degree of fracture healing. RESULTS Analysis showed complete secondary fracture healing in the control group without periosteal injury. Whereas the periosteal injury group demonstrated less than half as much maximum callus volume (p < 0.05) and bridging, recovery of stiffness and temporal expression of callus growth and remodelling was delayed by 7-15 days. CONCLUSION This paper introduces a novel mouse model of delayed union without a critically sized defect and with standardized biomechanical conditions, which enables further investigation into the molecular biological, biomechanical, and biochemical processes involved in (delayed) fracture healing and nonunion development. This model provides a continuum between normal fracture healing and the development of nonunions.
Collapse
|
12
|
Orth M, Altmeyer M, Scheuer C, Braun B, Holstein J, Eglin D, D'Este M, Histing T, Laschke M, Pohlemann T, Menger M. Effects of locally applied adipose tissue-derived microvascular fragments by thermoresponsive hydrogel on bone healing. Acta Biomater 2018; 77:201-211. [PMID: 30030175 DOI: 10.1016/j.actbio.2018.07.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/13/2018] [Accepted: 07/16/2018] [Indexed: 12/17/2022]
Abstract
Insufficient vascularization is a major cause for the development of non-unions. To overcome this problem, adipose tissue-derived microvascular fragments (MVF) may serve as vascularization units. However, their application into bone defects needs a carrier system. Herein, we analyzed whether this is achieved by a thermoresponsive hydrogel (TRH). MVF were isolated from CD-1 mice and cultivated after incorporation into TRH, while non-incorporated MVF served as controls. Viability of MVF was assessed immunohistochemically over a 7-day period. Moreover, osteotomies were induced in femurs of CD-1 mice. The osteotomy gaps were filled with MVF-loaded TRH (TRH + MVF), unloaded TRH (TRH) or no material (control). Bone healing was evaluated 14 and 35 days postoperatively. MVF incorporated into TRH exhibited less apoptotic cells and showed a stable vessel morphology compared to controls. Micro-computed tomography revealed a reduced bone volume in TRH + MVF femurs. Histomorphometry showed less bone and more fibrous tissue after 35 days in TRH + MVF femurs compared to controls. Accordingly, TRH + MVF femurs exhibited a lower osseous bridging score and a reduced bending stiffness. Histology and Western blot analysis revealed an increased vascularization and CD31 expression, whereas vascular endothelial growth factor (VEGF) expression was reduced in TRH + MVF femurs. Furthermore, the callus of TRH + MVF femurs showed increased receptor activator of NF-κB ligand expression and higher numbers of osteoclasts. These findings indicate that TRH is an appropriate carrier system for MVF. Application of TRH + MVF increases the vascularization of bone defects. However, this impairs bone healing, most likely due to lower VEGF expression during the early course of bone healing. STATEMENT OF SIGNIFICANCE In the present study we analyzed for the first time the in vivo performance of a thermoresponsive hydrogel (TRH) as a delivery system for bioactive microvascular fragments (MVF). We found that TRH represents an appropriate carrier for MVF as vascularization units and maintains their viability. Application of MVF-loaded TRH impaired bone formation in an established murine model of bone healing, although vascularization was improved. This unexpected outcome was most likely due to a reduced VEGF expression in the early phase bone healing.
Collapse
|
13
|
Williams JN, Li Y, Valiya Kambrath A, Sankar U. The Generation of Closed Femoral Fractures in Mice: A Model to Study Bone Healing. J Vis Exp 2018:58122. [PMID: 30176027 PMCID: PMC6128110 DOI: 10.3791/58122] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Bone fractures impose a tremendous socio-economic burden on patients, in addition to significantly affecting their quality of life. Therapeutic strategies that promote efficient bone healing are non-existent and in high demand. Effective and reproducible animal models of fractures healing are needed to understand the complex biological processes associated with bone regeneration. Many animal models of fracture healing have been generated over the years; however, murine fracture models have recently emerged as powerful tools to study bone healing. A variety of open and closed models have been developed, but the closed femoral fracture model stands out as a simple method for generating rapid and reproducible results in a physiologically relevant manner. The goal of this surgical protocol is to generate unilateral closed femoral fractures in mice and facilitate a post-fracture stabilization of the femur by inserting an intramedullary steel rod. Although devices such as a nail or a screw offer greater axial and rotational stability, the use of an intramedullary rod provides a sufficient stabilization for consistent healing outcomes without producing new defects in the bone tissue or damaging nearby soft tissue. Radiographic imaging is used to monitor the progression of callus formation, bony union, and subsequent remodeling of the bony callus. Bone healing outcomes are typically associated with the strength of the healed bone and measured with torsional testing. Still, understanding the early cellular and molecular events associated with fracture repair is critical in the study of bone tissue regeneration. The closed femoral fracture model in mice with intramedullary fixation serves as an attractive platform to study bone fracture healing and evaluate therapeutic strategies to accelerate healing.
Collapse
Affiliation(s)
- Justin N Williams
- Department of Anatomy and Cell Biology, Indiana University School of Medicine
| | - Yong Li
- Department of Anatomy and Cell Biology, Indiana University School of Medicine
| | | | - Uma Sankar
- Department of Anatomy and Cell Biology, Indiana University School of Medicine;
| |
Collapse
|
14
|
Histing T, Bremer P, Rollmann MF, Herath S, Klein M, Pohlemann T, Menger MD, Fritz T. A Minimally Invasive Model to Analyze Endochondral Fracture Healing in Mice Under Standardized Biomechanical Conditions. J Vis Exp 2018. [PMID: 29630050 DOI: 10.3791/57255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Bone healing models are necessary to analyze the complex mechanisms of fracture healing to improve clinical fracture treatment. During the last decade, an increased use of mouse models in orthopedic research was noted, most probably because mouse models offer a large number of genetically-modified strains and special antibodies for the analysis of molecular mechanisms of fracture healing. To control the biomechanical conditions, well-characterized osteosynthesis techniques are mandatory, also in mice. Here, we report on the design and use of a closed bone healing model to stabilize femur fractures in mice. The intramedullary screw, made of medical-grade stainless steel, provides through fracture compression an axial and rotational stability compared to the mostly used simple intramedullary pins, which show a complete lack of axial and rotational stability. The stability achieved by the intramedullary screw allows the analysis of endochondral healing. A large amount of callus tissue, received after stabilization with the screw, offers ideal conditions to harvest tissue for biochemical and molecular analyses. A further advantage of the use of the screw is the fact that the screw can be inserted into the femur with a minimally invasive technique without inducing damage to the soft tissue. In conclusion, the screw is a unique implant that can ideally be used in closed fracture healing models offering standardized biomechanical conditions.
Collapse
Affiliation(s)
- Tina Histing
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University;
| | - Philipp Bremer
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| | - Mika F Rollmann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| | - Steven Herath
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| | - Moritz Klein
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| | - Tim Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University
| | - Tobias Fritz
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| |
Collapse
|
15
|
Chen H, Sun X, Yin L, Chen S, Zhu Y, Huang J, Jiang W, Chen B, Zhang R, Chen L, Nie M, Xie Y, Deng Z. PTH 1-34 Ameliorates the Osteopenia and Delayed Healing of Stabilized Tibia Fracture in Mice with Achondroplasia Resulting from Gain-Of-Function Mutation of FGFR3. Int J Biol Sci 2017; 13:1254-1265. [PMID: 29104492 PMCID: PMC5666524 DOI: 10.7150/ijbs.21258] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/20/2017] [Indexed: 01/11/2023] Open
Abstract
Bone fracture healing is processed through multiple stages including the cartilaginous callus formation and its transition to bony callus. FGFR3 negatively regulates chondrogenesis and enhances osteogenesis during skeleton development. We previously found in mice carrying gain-of-function mutation of FGFR3 that FGFR3 delays the healing of un-stabilized fracture that heals mainly through endochondral ossification. Since fracture is regularly treated in clinics with rigid fixation, and stabilized fracture is healed largely through intramembranous ossification, we asked whether FGFR3, a key regulator of osteogenesis, also affect the regeneration of stabilized fracture. We found that gain-of-function mutation of FGFR3 inhibits the initiation of chondrogenesis and the subsequent bone formation. We further studied whether PTH1-34 can improve the osteopenia and delayed healing of the stabilized tibia fracture in mice with achondroplasia. Fracture healing was evaluated by radiography, micro-CT, biomechanical tests, histology, and real-time polymerase chain reaction (RT-PCR) analysis. We found that PTH 1-34 can alleviate the decreased bone mass and compromised architecture in ACH mice. Histological analysis revealed that administration of PTH1-34 increased the size of both the total callus and cartilaginous callus at 14 days after the surgery in ACH mice. RT-PCR data suggested that systemic PTH1-34 accelerated the initiation of chondrogenesis and chondrocyte maturation (earlier and higher levels of expression of chondrogenesis related markers) and enhanced the osteogenic differentiation in the fracture callus in ACH mice. These results indicate that the PTH1-34 administration resulted in an enhanced callus formation during bone fracture healing in ACH mice, which is at least in part mediated by an increase of cartilaginous callus at early stage and the promotion of bone formation in bony callus. In summary, in this study we revealed that FGFR3 delays the regeneration of stabilized fracture by inhibiting both the chondrogenesis and osteogenesis, and PTH1-34 treatment can improve the dysregulated bone metabolism and delayed bone injury healing resulting from gain-of-function mutation of FGFR3.
Collapse
Affiliation(s)
- Hangang Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China.,Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xianding Sun
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Liangjun Yin
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Shuai Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Ying Zhu
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Junlan Huang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Wanling Jiang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Bo Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Ruobin Zhang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Lin Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Mao Nie
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Yangli Xie
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zhongliang Deng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
16
|
Histing T, Menger MD, Pohlemann T, Matthys R, Fritz T, Garcia P, Klein M. An Intramedullary Locking Nail for Standardized Fixation of Femur Osteotomies to Analyze Normal and Defective Bone Healing in Mice. J Vis Exp 2016. [PMID: 27911364 DOI: 10.3791/54472] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Bone healing models are essential to the development of new therapeutic strategies for clinical fracture treatment. Furthermore, mouse models are becoming more commonly used in trauma research. They offer a large number of mutant strains and antibodies for the analysis of the molecular mechanisms behind the highly differentiated process of bone healing. To control the biomechanical environment, standardized and well-characterized osteosynthesis techniques are mandatory in mice. Here, we report on the design and use of an intramedullary nail to stabilize open femur osteotomies in mice. The nail, made of medical-grade stainless steel, provides high axial and rotational stiffness. The implant further allows the creation of defined, constant osteotomy gap sizes from 0.00 mm to 2.00 mm. Intramedullary locking nail stabilization of femur osteotomies with gap sizes of 0.00 mm and 0.25 mm result in adequate bone healing through endochondral and intramembranous ossification. Stabilization of femur osteotomies with a gap size of 2.00 mm results in atrophic non-union. Thus, the intramedullary locking nail can be used in healing and non-healing models. A further advantage of the use of the nail compared to other open bone healing models is the possibility to adequately fix bone substitutes and scaffolds in order to study the process of osseous integration. A disadvantage of the use of the intramedullary nail is the more invasive surgical procedure, inherent to all open procedures compared to closed models. A further disadvantage may be the induction of some damage to the intramedullary cavity, inherent to all intramedullary stabilization techniques compared to extramedullary stabilization procedures.
Collapse
Affiliation(s)
- Tina Histing
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University;
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University
| | - Tim Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| | | | - Tobias Fritz
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| | - Patric Garcia
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| | - Moritz Klein
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University
| |
Collapse
|
17
|
Fontaine JL, Hunt NA, Curry S, Kearney T, Jupiter D, Shibuya N, Lavery LA. Fracture healing and biomarker expression in a diabetic Zucker rat model. J Am Podiatr Med Assoc 2016; 104:428-33. [PMID: 25275729 DOI: 10.7547/0003-0538-104.5.428] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Persons with diabetes have a higher incidence of fractures compared with persons without diabetes. However, there is little published information concerning the deleterious effect of late-stage diabetes on fracture healing. There are no studies using animal models that evaluate the effect of advanced diabetes on fracture healing. The purpose of our study was to evaluate cytokine expression, specifically macrophage inflammatory protein 1 (MIP-1) and vascular endothelial growth factor, in fracture healing in a type 2 diabetes rat model. METHODS We evaluated biomarker expression after femur fracture using a rat model. The two groups consisted of 24 Zucker diabetic rats (study group) and 12 Zucker lean rats (control group). An independent reviewer was used to assess delayed union. We evaluated serum samples 2, 4, 7, and 14 days after surgery for MIP-1, vascular endothelial growth factor, leptin, and other cytokine levels. RESULTS At 3 weeks, Kaplan-Meier estimates showed that 45.8% of femur fractures in Zucker diabetic rats had healed, whereas 81.8% of those in Zucker lean rats had healed (P = .02). A logistic regression model to predict fast healing that included the three cytokines and diabetes status showed that the only factor achieving significance was MIP-1α. Vascular endothelial growth factor was the only biomarker to show significance compared with delayed healing. CONCLUSIONS These results confirm significant differences in biomarker expression between diabetic and nondiabetic rats during bone healing. The key factors for bone healing may appear early in the healing process, whereas differences in diabetes versus nondiabetes are seen later in the healing process. Increased levels of MIP-1α were associated with the likelihood of delayed healing.
Collapse
Affiliation(s)
- Javier La Fontaine
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Nathan A. Hunt
- Orthopaedic and Spine Center of the Rockies, Fort Collins, CO
| | - Stacey Curry
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | | | - Daniel Jupiter
- Department of Surgery, Texas A&M Health and Science Center College of Medicine and Scott and White Memorial Hospital and Clinics, Central Texas Veterans Affairs Health Care System, Temple, TX
| | - Naohiro Shibuya
- Department of Surgery/Podiatry, Central Texas VA Health Care System, Temple, TX
| | - Lawrence A. Lavery
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
18
|
Hagiwara Y, Dyment N, Jiang X, Huang J, Ackert-Bicknell C, Adams D, Rowe D. Fixation stability dictates the differentiation pathway of periosteal progenitor cells in fracture repair. J Orthop Res 2015; 33:948-56. [PMID: 25639792 PMCID: PMC4891973 DOI: 10.1002/jor.22816] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 12/02/2014] [Accepted: 12/19/2014] [Indexed: 02/04/2023]
Abstract
This study compared fracture repair stabilized by intramedullary pin (IMP) or external fixation (EF) in GFP reporter mice. A modified IMP was used as control while EF utilized six needles inserted transversely through the tibia and into a segment of a syringe barrel. X-rays taken at days 0, 14, and 35 showed that IMP resulted in significant three-dimensional deformity with a large callus while EF showed minimal deformity and callus formation. Cryohistological analysis of IMP at day 14 confirmed a large ColX-RFPchry+ callus surrounded by woven bone (Col3.6-GFPcyan) and TRAP+ osteoclasts with mature bone (hOC-GFPtpz) at the base. By day 35, cartilaginous components had been resorbed and an outer cortical shell (OCS) showed evidence of inward modeling. In contrast, the EF at day 14 showed no evidence of cartilage formation. Instead, periosteal-derived osteoblasts (Col3.6-GFPcyan) entered the fracture cleft and formed woven bone that spanned the marrow space. By day 35, mature bone had formed that was contiguous with the opposing cortical bone. Fracture site stability greatly affects the cellular response during repair and must be considered in the preclinical models that test therapies for improving fracture healing.
Collapse
Affiliation(s)
- Y. Hagiwara
- Department of Orthopedic Surgery, Nippon Medical School Hospital, Tokyo 113, JAPAN
| | | | | | | | - C. Ackert-Bicknell
- Dept. Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester, NY 14642
| | - D.J. Adams
- Department of Orthopaedic Surgery, New England Musculoskeletal Institute, University of Connecticut Health Center, Farmington, CT 06030
| | | |
Collapse
|
19
|
Furuya H, Tabata Y, Kaneko K. Bone Regeneration for Murine Femur Fracture by Gelatin Hydrogels Incorporating Basic Fibroblast Growth Factor with Different Release Profiles. Tissue Eng Part A 2014; 20:1531-41. [DOI: 10.1089/ten.tea.2012.0763] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Hiroyuki Furuya
- Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yasuhiko Tabata
- Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kazuo Kaneko
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Zhao X, Wang JX, Feng YF, Wu ZX, Zhang Y, Shi L, Tan QC, Yan YB, Lei W. Systemic treatment with telmisartan improves femur fracture healing in mice. PLoS One 2014; 9:e92085. [PMID: 24642982 PMCID: PMC3958447 DOI: 10.1371/journal.pone.0092085] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/17/2014] [Indexed: 11/29/2022] Open
Abstract
Recent clinical studies indicated that angiotensin receptor blockers (ARBs) would decrease the risk of bone fractures in the elderly populations. There is little known about the role of the ARBs in the process of fracture healing. The purpose of the present study was to verify the hypothesis that systemic treatment with telmisartan has the ability to promote fracture healing. In this study, femur fractures were produced in 96 mature male BALB/c mice. Animals were treated with the ARBs telmisartan or vehicle. Fracture healing was analysed after 2, 5 and 10 weeks postoperatively using X-ray, biomechanical testing, histomorphometry, immunohistochemistry and micro-computed tomography (micro-CT). Radiological analysis showed the diameter of the callus in the telmisartan treated animals was significantly increased when compared with that of vehicle treated controls after two weeks of fracture healing. The radiologically observed promotion of callus formation was confirmed by histomorphometric analyses, which revealed a significantly increased amount of bone formation when compared with vehicle-treated controls. Biomechanical testing further showed a significantly greater peak torque at failure, and a higher torsional stiffness in telmisartan-treated animals compared with controls. There was an increased fraction of PCNA-positive cells and VEGF-positive cells in telmisartan-treated group compared with vehicle-treated controls. From the three-dimensional reconstruction of the bony callus, telmisartan-treated group significantly increased the values of BV/TV by 21.7% and CsAr by 26.0% compared to the vehicle-treated controls at 5 weeks post-fracture. In summary, we demonstrate in the current study that telmisartan could promote fracture healing in a mice model via increasing mechanical strength and improving microstructure. The most mechanism is probably by an increase of cell proliferation and neovascularization associated with a decreased VEGF expression in hypertrophic chondrocytes.
Collapse
Affiliation(s)
- Xiong Zhao
- Department of Orthopeadics, Xijing Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Jia-xing Wang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, PR China
- ICU, 309th Hospital of PLA, Beijing, PR China
| | - Ya-fei Feng
- Department of Orthopeadics, Xijing Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Zi-xiang Wu
- Department of Orthopeadics, Xijing Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Yang Zhang
- Department of Orthopeadics, Xijing Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Lei Shi
- Department of Orthopeadics, Xijing Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Quan-chang Tan
- Department of Orthopeadics, Xijing Hospital, The Fourth Military Medical University, Xi'an, PR China
| | - Ya-bo Yan
- Department of Orthopeadics, Xijing Hospital, The Fourth Military Medical University, Xi'an, PR China
- * E-mail: (WL); (YY)
| | - Wei Lei
- Department of Orthopeadics, Xijing Hospital, The Fourth Military Medical University, Xi'an, PR China
- * E-mail: (WL); (YY)
| |
Collapse
|
21
|
Eardley WGP, Watts SA, Clasper JC. Modelling for conflict: the legacy of ballistic research and current extremity in vivo modelling. J ROY ARMY MED CORPS 2013; 159:73-83. [PMID: 23720587 DOI: 10.1136/jramc-2013-000074] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Extremity ballistic injury is unique and the literature intended to guide its management is commonly misinterpreted. In order to care for those injured in conflict and conduct appropriate research, clinicians must be able to identify key in vivo studies, understand their weaknesses and desist the propagation of miscited and misunderstood ballistic dogma. This review provides the only inclusive critical overview of key studies of relevance to military extremity injury. In addition, the non-ballistic studies of limb injury, stabilisation and contamination that will form the basis from which future small animal extremity studies are constructed are presented. With an awareness of the legacy of military wound models and an insight into available generic models of extremity injury and contamination, research teams are well placed to optimise future military extremity injury management.
Collapse
Affiliation(s)
- William G P Eardley
- Academic Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, ICT Centre, Institute of Research and Development, Birmingham, UK.
| | | | | |
Collapse
|
22
|
Liu K, Li D, Huang X, Lv K, Ongodia D, Zhu L, Zhou L, Li Z. A murine femoral segmental defect model for bone tissue engineering using a novel rigid internal fixation system. J Surg Res 2013; 183:493-502. [PMID: 23522461 DOI: 10.1016/j.jss.2013.02.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/04/2013] [Accepted: 02/20/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND As a model animal, the mouse has already been widely used in bone-related research. However, there is a lack of ideal long bone segmental defect mouse model. Since external fixation has disadvantages of heavy weight, penetrating the skin, and hampering mobility, an internal fixation device is probably more preferable to maintain the segmental bone defect. The aim of this study was to establish a simple, reproducible, and standardized murine critical-size defect model through designing an internal fixation system, verifying its adaptability, and investigating the critical size of femoral segmental defect. METHODS By utilizing computer-aided measuring and processing system, anatomical data of adult C57BL/6 mouse femur was obtained, and a plate-bolts system was designed for rigid fixation. The plate and screws were fixed in 67 mice and 1.5 or 2.0 mm defect gaps were created in the femoral midshaft. Compression and three-point bending of bone-implant construct were tested in mice at 0, 2, 5, and 12 wk postoperative to test the biomechanical stability. X-ray, micro-computed tomography, and histology were used to investigate the defect healing process. RESULTS The plate- and screws-fitted mouse femur and unilateral or bilateral operation had seemingly no adverse impact on the mouse in general. Mechanical tests indicated that there were no significant differences between the bone-implant construct and intact femur in compression and three-point bending loading. Micro-computed tomography scanning showed the bone mineral density had not been affected by the implantation of fixation device. There was no union of the 2.0 mm segmental defect in 12-wk period. CONCLUSION Using the specifically designed rigid internal fixation device, a segmental defect size of 2.0 mm in C57BL/6 mouse femur will show nonunion and can serve as a critical defect size for bone tissue engineering and bone regeneration research.
Collapse
Affiliation(s)
- Kai Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Manassero M, Viateau V, Matthys R, Deschepper M, Vallefuoco R, Bensidhoum M, Petite H. A novel murine femoral segmental critical-sized defect model stabilized by plate osteosynthesis for bone tissue engineering purposes. Tissue Eng Part C Methods 2012; 19:271-80. [PMID: 22953787 DOI: 10.1089/ten.tec.2012.0256] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mouse models are invaluable tools for mechanistic and efficacy studies of the healing process of large bone defects resulting in atrophic nonunions, a severe medical problem and a financial health-care-related burden. Models of atrophic nonunions are usually achieved by providing a highly stable biomechanical environment. For this purpose, external fixators have been investigated, but plate osteosynthesis, despite its high clinical relevance, has not yet been considered in mice. We hereby proposed and investigated the use of an internal osteosynthesis for stabilizing large bone defects. To this aim, a 3.5-mm-long segmental bone defect was induced in the mid-shaft of the femur using a Gigli saw and a jig. Bone fixation was performed using a titanium microlocking plate with four locking screws. The bone defect was either left empty or filled with a syngenic bone graft or filled with a coralline scaffold. Healing was monitored using radiographs. The healing process was further assessed using microcomputed tomography and histology 10 weeks after surgery. With the exception of one mouse that died during the surgical procedure, no complications were observed. A stable and reproducible bone fixation as well as a reproducible fixation of the implanted materials with full weight bearing was obtained in all animals tested. Nonunion was consistently observed in the group in which the defects were left empty. Bone union was obtained with the syngenic bone grafts, providing evidence that, although such defects were of critical size, bone healing was possible when the gold-standard material was used to fill the defect. Although new bone formation was greater in the coralline scaffold group than in the left-empty animal group, it remained limited and localized close to the bony edges, a consequence of the critical size of such bone defect. Our study established a reproducible, clinically relevant, femoral, atrophic nonunion, critical-sized defect, low morbidity mouse model. The present study was successful in designing and testing in a small animal model, a novel surgical method for the assessment of bone repair; this model has the potential to facilitate investigations of the molecular and cellular events involved in bone regeneration in load-bearing, segmental-bone defects.
Collapse
Affiliation(s)
- Mathieu Manassero
- Laboratory of Bioengineering and Biomechanics for Bone Articulation (B2OA–UMR CNRS 7052), University Paris-Diderot, Paris, France
| | | | | | | | | | | | | |
Collapse
|
24
|
Melatonin Impairs Fracture Healing by Suppressing RANKL-Mediated Bone Remodeling. J Surg Res 2012; 173:83-90. [DOI: 10.1016/j.jss.2010.08.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Revised: 08/09/2010] [Accepted: 08/19/2010] [Indexed: 11/23/2022]
|
25
|
Garcia P, Pieruschka A, Klein M, Tami A, Histing T, Holstein JH, Scheuer C, Pohlemann T, Menger MD. Temporal and spatial vascularization patterns of unions and nonunions: role of vascular endothelial growth factor and bone morphogenetic proteins. J Bone Joint Surg Am 2012; 94:49-58. [PMID: 22218382 DOI: 10.2106/jbjs.j.00795] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Failure of fracture-healing with nonunion is a major clinical problem. Angiogenesis is closely linked to bone regeneration, but the role of angiogenesis in nonunion formation remains unclear. Because established nonunions are well vascularized, we hypothesized that lack of vascular endothelial growth factor (VEGF) expression and vascularization during the early time course of fracture-healing determine nonunion formation. METHODS In seventy-two CD-1 mice, a femoral osteotomy with a gap size of 1.80 mm (nonunion group) or a gap size of 0.25 mm (union group) was created and stabilized by a pin-clip technique. Healing was analyzed after three, seven, fourteen, twenty-one, twenty-eight, and seventy days by micro-computed tomography and histomorphometry. Vascularization was determined in different healing zones by immunohistochemical staining of PECAM-1 (platelet-endothelial cell adhesion molecule). Additional animals were analyzed after seven, fourteen, and twenty-one days with Western blot analysis of VEGF, bone morphogenetic protein (BMP)-2, and BMP-4 expression. RESULTS Micro-computed tomography and histomorphometry showed complete bone-bridging in the union group, whereas animals in the nonunion group showed atrophic nonunion formation. Vascularization increased from day 3 to day 7 in both groups, with a subsequent decrease after fourteen days. However, overall vascularization did not differ between unions and nonunions over time. It is of interest that vascularization within the endosteal healing zone was even higher in nonunions than in unions after fourteen days. Expression of VEGF was significantly higher in nonunions, while expression of BMP-2 and 4 and proliferating cell nuclear antigen were found significantly reduced compared with unions. CONCLUSIONS Because vascularization during the early time course of fracture-healing was not impaired despite the failure of bone-healing in nonunions, we rejected our hypothesis and accepted the null hypothesis that nonunion formation is not due to failure of VEGF-mediated angiogenesis. Failure of fracture-healing was associated with a decreased expression of BMP-2 and 4 and a disturbed ratio of angiogenic to osteogenic growth factors, which may be responsible for nonunion. CLINICAL RELEVANCE Because the intrinsic angiogenic response during nonunion formation was sufficient for adequate vascularization, treatment strategies for nonunions should focus on the stimulation of osteogenesis rather than on the stimulation of angiogenesis.
Collapse
Affiliation(s)
- P Garcia
- Department of Trauma, Hand and Reconstructive Surgery, Collaborative Research Center, AO Foundation, University of Saarland, D-66421 Homburg/Saar, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tavassol F, Kampmann A, Schumann P, Lindhorst D, Kokemüller H, Essig H, Meemken JH, Rücker M, Gellrich NC. A Novel Approach for Studying Microcirculation in Bone Defects by Intravital Fluorescence Microscopy. Tissue Eng Part C Methods 2011; 17:1151-9. [DOI: 10.1089/ten.tec.2011.0065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Frank Tavassol
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Andreas Kampmann
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Paul Schumann
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Daniel Lindhorst
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Horst Kokemüller
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Harald Essig
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Jan-Helge Meemken
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Martin Rücker
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Nils-Claudius Gellrich
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
27
|
Holstein JH, Orth M, Scheuer C, Tami A, Becker SC, Garcia P, Histing T, Mörsdorf P, Klein M, Pohlemann T, Menger MD. Erythropoietin stimulates bone formation, cell proliferation, and angiogenesis in a femoral segmental defect model in mice. Bone 2011; 49:1037-45. [PMID: 21851867 DOI: 10.1016/j.bone.2011.08.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 08/02/2011] [Accepted: 08/02/2011] [Indexed: 11/23/2022]
Abstract
The glycoprotein erythropoietin (EPO) has been demonstrated to stimulate fracture healing. The aim of the present study was to investigate the effect of EPO treatment on bone repair in a femoral segmental defect model. Bone repair was analyzed in mice which were treated by EPO (500IE/kg/d intraperitoneally; n=38) and in mice which received the vehicle for control (n=40). Two and 10 weeks after creating a 1.8mm femoral segmental defect, bone repair was studied by micro-CT, histology, and Western blot analysis. At 10 weeks, micro-CT and histomorphometric analyses showed a significantly higher bridging rate of the bone defects in EPO-treated animals than in controls. This was associated by a significantly higher bone volume within the segmental defects of the EPO-treated animals. At 2 weeks, Western blot analyses revealed a significantly higher expression of vascular endothelial growth factor (VEGF) in EPO-treated animals compared to controls. Accordingly, the number of blood vessels was significantly increased in the EPO group at 2 weeks. At 10 weeks, we found a significantly higher expression of proliferating cell nuclear antigen (PCNA) in EPO-treated animals when compared to controls. Western blot analyses showed no significant differences between the groups in the expression of the endothelial and inducible nitric oxide synthases (eNOS and iNOS) and the angiopoietin receptor Tie-2. Immunohistochemistry confirmed the results of the Western blot analyses, demonstrating a significantly higher number of VEGF- and PCNA-positive cells in EPO-treated animals than in controls at 2 and 10 weeks, respectively. We conclude that EPO is capable of stimulating bone formation, cell proliferation and VEGF-mediated angiogenesis in a femoral segmental defect model.
Collapse
Affiliation(s)
- J H Holstein
- Department of Trauma, Hand & Reconstructive Surgery, University of Saarland, Kirrberger Strasse 1, 66421 Homburg/Saar, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Histing T, Garcia P, Holstein JH, Klein M, Matthys R, Nuetzi R, Steck R, Laschke MW, Wehner T, Bindl R, Recknagel S, Stuermer EK, Vollmar B, Wildemann B, Lienau J, Willie B, Peters A, Ignatius A, Pohlemann T, Claes L, Menger MD. Small animal bone healing models: standards, tips, and pitfalls results of a consensus meeting. Bone 2011; 49:591-9. [PMID: 21782988 DOI: 10.1016/j.bone.2011.07.007] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Revised: 07/02/2011] [Accepted: 07/05/2011] [Indexed: 01/26/2023]
Abstract
Small animal fracture models have gained increasing interest in fracture healing studies. To achieve standardized and defined study conditions, various variables must be carefully controlled when designing fracture healing experiments in mice or rats. The strain, age and sex of the animals may influence the process of fracture healing. Furthermore, the choice of the fracture fixation technique depends on the questions addressed, whereby intra- and extramedullary implants as well as open and closed surgical approaches may be considered. During the last few years, a variety of different, highly sophisticated implants for fracture fixation in small animals have been developed. Rigid fixation with locking plates or external fixators results in predominantly intramembranous healing in both mice and rats. Locking plates, external fixators, intramedullary screws, the locking nail and the pin-clip device allow different degrees of stability resulting in various amounts of endochondral and intramembranous healing. The use of common pins that do not provide rotational and axial stability during fracture stabilization should be discouraged in the future. Analyses should include at least biomechanical and histological evaluations, even if the focus of the study is directed towards the elucidation of molecular mechanisms of fracture healing using the largely available spectrum of antibodies and gene-targeted animals to study molecular mechanisms of fracture healing. This review discusses distinct requirements for the experimental setups as well as the advantages and pitfalls of the different fixation techniques in rats and mice.
Collapse
Affiliation(s)
- T Histing
- Department of Trauma, Hand and Reconstructive Surgery, University of Saarland, Homburg/Saar, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Steck R, Ueno M, Gregory L, Rijken N, Wullschleger ME, Itoman M, Schuetz MA. Influence of internal fixator flexibility on murine fracture healing as characterized by mechanical testing and microCT imaging. J Orthop Res 2011; 29:1245-50. [PMID: 21437963 DOI: 10.1002/jor.21341] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Accepted: 11/29/2010] [Indexed: 02/04/2023]
Abstract
Mechanically well-defined stabilization systems have only recently become available, providing standardized conditions for studying the role of the mechanical environment on mouse bone fracture healing. The aim of this study was to characterize the time course of strength recovery and callus development of mouse femoral osteotomies stabilized with either low or high flexibility (in bending and torsion) internal fixation plates. Animals were euthanized and femora excised at 14, 21, and 28 days post-osteotomy for microCT analysis and torsional strength testing. While a larger mineralized callus was observed in osteotomies under more flexible conditions at all time points, the earlier bridging of the mineralized callus under less flexible conditions by 1 week resulted in an earlier recovery of torsional strength in mice stabilized with low flexibility fixation. Ultimate torque values for these bones were significantly higher at 14 and 21 days post-osteotomy compared to bones with the more flexible stabilization. Our study confirms the high reproducibility of the results that are achieved with this new implant system, therefore making it ideal for studying the influence of the mechanical environment on murine fracture healing under highly standardized conditions.
Collapse
Affiliation(s)
- Roland Steck
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Preclinical modeling of human disease with animals has not been standardized for many common pathologic processes. Assorted animal models are being used to investigate the pathogenesis, prevention, and treatment of disease processes. Certainly it is difficult to interpret the current literature because there are diverse and often irrelevant models being implemented. Some models are used for reasons of size or ease rather than the true modeling of a physiological process. Application to granting agencies and design of animal studies is difficult without standardization of the ideal preclinical model for disease states. The current article addresses the preclinical animal modeling of osteoporosis, infection, bone defects, and cartilage injury. This article is a discussion of the current literature, commonly used models, and suggests preferred preclinical models for future research design.
Collapse
|
31
|
The LockingMouseNail—A New Implant for Standardized Stable Osteosynthesis in Mice. J Surg Res 2011; 169:220-6. [DOI: 10.1016/j.jss.2009.11.713] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 11/05/2009] [Accepted: 11/13/2009] [Indexed: 11/20/2022]
|
32
|
Histing T, Marciniak K, Scheuer C, Garcia P, Holstein JH, Klein M, Matthys R, Pohlemann T, Menger MD. Sildenafil accelerates fracture healing in mice. J Orthop Res 2011; 29:867-73. [PMID: 21246617 DOI: 10.1002/jor.21324] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 11/08/2010] [Indexed: 02/04/2023]
Abstract
Sildenafil, a cyclic guanosine monophosphate (cGMP)-dependent phospodiesterase-5 inhibitor, has been shown to be a potent stimulator of angiogenesis through upregulation of pro-angiogenic factors and control of cGMP concentration. Herein, we determined whether sildenafil also influences angiogenic growth factor expression and bone formation during the process of fracture healing. Bone healing was studied in a murine closed femur fracture model using radiological, biomechanical, histomorphometric, and protein biochemical analysis at 2 and 5 weeks after fracture. Thirty mice received 5 mg/kg body weight sildenafil p.o. daily. Controls (n = 30) received equivalent amounts of vehicle. After 2 weeks of fracture healing sildenafil significantly increased osseous fracture bridging, as determined radiologically and histologically. This resulted in an increased biomechanical stiffness compared to controls. A smaller callus area with a slightly reduced amount of cartilaginous tissue indicated an accelerated healing process. After 5 weeks the differences were found blunted, demonstrating successful healing in both groups. Western blot analysis showed a significantly higher expression of the pro-angiogenic and osteogenic cysteine-rich protein (CYR) 61, confirming the increase of bone formation. We show for the first time that sildenafil treatment accelerates fracture healing by enhancing bone formation, most probably by a CYR61-associated pathway.
Collapse
Affiliation(s)
- Tina Histing
- Department of Trauma, Hand and Reconstructive Surgery, University of Saarland, Homburg/Saar, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Gardner MJ, Putnam S, Wong A, Streubel PN, Kotiya A, Silva MJ. Differential fracture healing resulting from fixation stiffness variability: a mouse model. J Orthop Sci 2011; 16:298-303. [PMID: 21451972 PMCID: PMC3580844 DOI: 10.1007/s00776-011-0051-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 01/21/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND The mechanisms underlying the interaction between the local mechanical environment and fracture healing are not known. We developed a mouse femoral fracture model with implants of different stiffness, and hypothesized that differential fracture healing would result. METHODS Femoral shaft fractures were created in 70 mice, and were treated with an intramedullary nail made of either tungsten (Young's modulus = 410 GPa) or aluminium (Young's modulus = 70 GPa). Mice were then sacrificed at 2 or 5 weeks. Fracture calluses were analyzed using standard microCT, histological, and biomechanical methods. RESULTS At 2 weeks, callus volume was significantly greater in the aluminium group than in the tungsten group (61.2 vs. 40.5 mm(3), p = 0.016), yet bone volume within the calluses was no different between the groups (13.2 vs. 12.3 mm(3)). Calluses from the tungsten group were stiffer on mechanical testing (18.7 vs. 9.7 N/mm, p = 0.01). The percent cartilage in the callus was 31.6% in the aluminium group and 22.9% in the tungsten group (p = 0.40). At 5 weeks, there were no differences between any of the healed femora. CONCLUSIONS In this study, fracture implants of different stiffness led to different fracture healing in this mouse fracture model. Fractures treated with a stiffer implant had more advanced healing at 2 weeks, but still healed by callus formation. Although this concept has been well documented previously, this particular model could be a valuable research tool to study the healing consequences of altered fixation stiffness, which may provide insight into the pathogenesis and ideal treatment of fractures and non-unions.
Collapse
|
34
|
An innovative intramedullary nail for stabilizing mouse femoral defects. J Surg Res 2011; 167:39-40. [PMID: 20828755 DOI: 10.1016/j.jss.2010.05.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 04/28/2010] [Accepted: 05/24/2010] [Indexed: 10/19/2022]
|
35
|
Garcia P, Speidel V, Scheuer C, Laschke MW, Holstein JH, Histing T, Pohlemann T, Menger MD. Low dose erythropoietin stimulates bone healing in mice. J Orthop Res 2011; 29:165-72. [PMID: 20740668 DOI: 10.1002/jor.21219] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Accepted: 06/18/2010] [Indexed: 02/04/2023]
Abstract
Beyond its classical role in regulation of erythropoiesis, erythropoietin (EPO) has been shown to exert protective and regenerative actions in a variety of non-hematopoietic tissues. However, little is known about potential actions in bone regeneration. To analyze fracture healing in mice, a femoral 0.25 mm osteotomy gap was stabilized with a pin-clip technique. Animals were treated with 500 U EPO/kg bw per day or with vehicle only. After 2 and 5 weeks, fracture healing was analyzed biomechanically, radiologically and histologically. Expression of PCNA and NFκB was examined by Western blot analysis. Vascularization was analyzed by immunohistochemical staining of PECAM-1. Circulating endothelial progenitor cells were measured by flow-cytometry. Herein, we demonstrate that EPO-treatment significantly accelerates bone healing in mice. This is indicated by a significantly greater biomechanical stiffness and a higher radiological density of the periosteal callus at 2 and 5 weeks after fracture and stabilization. Histological analysis demonstrated significantly more bone and less cartilage and fibrous tissue in the periosteal callus. Endosteal vascularization was significantly increased in EPO-treated animals when compared to controls. The number of circulating endothelial progenitor cells was significantly greater in EPO-treated animals. The herein shown acceleration of healing by EPO may represent a promising novel treatment strategy for fractures with delayed healing and non-union formation.
Collapse
Affiliation(s)
- P Garcia
- Department of Trauma-, Hand- and Reconstructive Surgery, University of Saarland, Homburg/Saar, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Experimental trauma models: an update. J Biomed Biotechnol 2011; 2011:797383. [PMID: 21331361 PMCID: PMC3035380 DOI: 10.1155/2011/797383] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Accepted: 12/17/2010] [Indexed: 01/31/2023] Open
Abstract
Treatment of polytrauma patients remains a medical as well as socioeconomic challenge. Although diagnostics and therapy improved during the last decades, multiple injuries are still the major cause of fatalities in patients below 45 years of age. Organ dysfunction and organ failure are major complications in patients with major injuries and contribute to mortality during the clinical course. Profound understanding of the systemic pathophysiological response is crucial for innovative therapeutic approaches. Therefore, experimental studies in various animal models are necessary. This review is aimed at providing detailed information of common trauma models in small as well as in large animals.
Collapse
|
37
|
Histing T, Kristen A, Roth C, Holstein J, Garcia P, Matthys R, Menger M, Pohlemann T. In vivo gait analysis in a mouse femur fracture model. J Biomech 2010; 43:3240-3. [DOI: 10.1016/j.jbiomech.2010.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2009] [Revised: 06/25/2010] [Accepted: 07/24/2010] [Indexed: 11/25/2022]
|
38
|
Röntgen V, Blakytny R, Matthys R, Landauer M, Wehner T, Göckelmann M, Jermendy P, Amling M, Schinke T, Claes L, Ignatius A. Fracture healing in mice under controlled rigid and flexible conditions using an adjustable external fixator. J Orthop Res 2010; 28:1456-62. [PMID: 20872581 DOI: 10.1002/jor.21148] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mice are increasingly used to investigate mechanobiology in fracture healing. The need exists for standardized models allowing for adjustment of the mechanical conditions in the fracture gap. We introduced such a model using rigid and flexible external fixators with considerably different stiffness (axial stiffnesses of 18.1 and 0.82 N/mm, respectively). Both fixators were used to stabilize a 0.5 mm osteotomy gap in the femur of C57BL/6 mice (each n = 8). Three-point bending tests, µCT, and histomorphometry demonstrated a different healing pattern after 21 days. Both fixations induced callus formation with a mixture of intramembranous and enchondral ossification. Under flexible conditions, the bending stiffness of the callus was significantly reduced, and a larger but qualitatively inferior callus with a significantly lower fraction of bone but a higher fraction of cartilage and soft tissue was formed. Monitoring of the animal movement and the ground reaction forces demonstrated physiological loading with no significant differences between the groups, suggesting that the differences in healing were not based on a different loading behavior. In summary, flexible external fracture fixation of the mouse femur led to delayed fracture healing in comparison to a more rigid situation.
Collapse
Affiliation(s)
- Viktoria Röntgen
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Garcia P, Schwenzer S, Slotta JE, Scheuer C, Tami AE, Holstein JH, Histing T, Burkhardt M, Pohlemann T, Menger MD. Inhibition of angiotensin-converting enzyme stimulates fracture healing and periosteal callus formation - role of a local renin-angiotensin system. Br J Pharmacol 2010; 159:1672-80. [PMID: 20233225 DOI: 10.1111/j.1476-5381.2010.00651.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND AND PURPOSE The renin-angiotensin system (RAS) regulates blood pressure and electrolyte homeostasis. In addition, 'local' tissue-specific RAS have been identified, regulating regeneration, cell growth, apoptosis, inflammation and angiogenesis. Although components of the RAS are expressed in osteoblasts and osteoclasts, a local RAS in bone has not yet been described and there is no information on whether the RAS is involved in fracture healing. Therefore, we studied the expression and function of the key RAS component, angiotensin-converting enzyme (ACE), during fracture healing. EXPERIMENTAL APPROACH In a murine femur fracture model, animals were treated with the ACE inhibitor perindopril or vehicle only. Fracture healing was analysed after 2, 5 and 10 weeks using X-ray, micro-CT, histomorphometry, immunohistochemistry, Western blotting and biomechanical testing. KEY RESULTS ACE was expressed in osteoblasts and hypertrophic chondrocytes in the periosteal callus during fracture healing, accompanied by expression of the angiotensin type-1 and type-2 receptors. Perindopril treatment reduced blood pressure and bone mineral density in unfractured femora. However, it improved periosteal callus formation, bone bridging of the fracture gap and torsional stiffness. ACE inhibition did not affect cell proliferation, but reduced apoptotic cell death. After 10 week treatment, a smaller callus diameter and bone volume after perindopril treatment indicated an advanced stage of bone remodelling. CONCLUSIONS Our study provides evidence for a local RAS in bone that influenced the process of fracture healing. We show for the first time that inhibition of ACE is capable of accelerating bone healing and remodelling.
Collapse
Affiliation(s)
- P Garcia
- Department of Trauma-, Hand- and Reconstructive Surgery, University of Saarland, Homburg/Saar, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Advances in the establishment of defined mouse models for the study of fracture healing and bone regeneration. J Orthop Trauma 2009; 23:S31-8. [PMID: 19390374 DOI: 10.1097/bot.0b013e31819f27e5] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The availability of a broad spectrum of antibodies and gene-targeted animals caused an increasing interest in mouse models for the study of molecular mechanisms of fracture healing and bone regeneration. In most murine fracture models, the tibia or the femur is fractured using a 3-point bending device (closed models) or is osteotomized using an open surgical approach (open models). For fracture studies in mice, the tibia has to be considered less appropriate compared with the femur because the stabilization of the fracture is more difficult due to its triangular, distally declining caliber and its bowed longitudinal axis. Biomechanical factors critically influence the bone healing process. Thus, the use of stable osteosynthesis techniques is also of interest in murine fracture models. To achieve stable fixation, several biomechanically standardized implants have recently been introduced, including a locking nail and an intramedullary compression screw. Other implants, such as a pin-clip, an external fixator, and a locking plate, additionally allow the stabilization of fractures with distinct gap sizes. This enables the study of healing of critical size defects and nonunions. The use of these implants further allows a rigid fixation of fractures in bridle bones, which is essential for fracture studies in animals suffering from metabolic bone diseases like osteoporosis. In general, the analysis of bone healing in these models includes different imaging techniques and histologic, immunohistochemical, biomechanical, and molecular methods. To evaluate the impact of different osteosynthesis techniques on physical activity and rehabilitation, gait analysis may additionally be performed. By this, the gait of the animals can be visualized and quantitatively analyzed using modified running wheels and dynamic high-resolution radiography systems. Taken together, a variety of different murine femur fracture models have become available, providing defined biomechanical conditions for fracture research. The use of these mouse models may now allow studying the influence of fracture stabilization techniques on molecular mechanisms of bone healing.
Collapse
|