Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 19(9)2018 Aug 25.
Article in English | MEDLINE | ID: mdl-30149650

ABSTRACT

The application of autologous mesenchymal stem cells (MSC) for the treatment of bone defects requires two invasive procedures and several weeks of ex vivo cell expansion. To overcome these limitations, the administration of allogeneic MSC may be attractive, because they are anticipated to be immunoprivileged. Because preclinical studies using various animal models are conflicting with respect to the efficacy of allogeneic MSC, we investigated whether autologous and allogeneic human MSC (hMSC) are equally effective in regenerating bone in a humanized mouse model resembling the human immune system. Applying autologous and allogeneic hMSC in critically sized femoral defects, we found that allogeneic hMSC elicited a mild immune response early after implantation, whereas early angiogenic processes were similar in both treatments. At later healing time points, the transplantation of allogeneic hMSC resulted in less bone formation than autologous hMSC, associated with a reduced expression of the osteogenic factor Runx2 and impaired angiogenesis. We found by species-specific staining for collagen-type-1α2 that MSCs of either source did not synthesize new bone matrix, indicating an indirect contribution of transplanted hMSC to bone regeneration. In conclusion, our data suggest that the application of autologous hMSC is superior to that of allogeneic cells for bone defect treatment.


Subject(s)
Bone Regeneration , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Humans , Immunity, Cellular , Mesenchymal Stem Cells/immunology , Mice , Mice, Transgenic , Neovascularization, Physiologic , Osteogenesis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Time Factors , Transplantation, Autologous , Transplantation, Homologous , Wound Healing
2.
Acta Orthop ; 88(2): 217-222, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27841708

ABSTRACT

Background and purpose - Constant fixator stiffness for the duration of healing may not provide suitable mechanical conditions for all stages of bone repair. We therefore investigated the influence of stiffening fixation on callus stiffness and morphology in a rat diaphyseal osteotomy model to determine whether healing time was shortened and callus stiffness increased through modulation of fixation from flexible to stiff. Material and methods - An external unilateral fixator was applied to the osteotomized femur and stiffened by decreasing the offset of the inner fixator bar at 3, 7, 14, and 21 days after operation. After 5 weeks, the rats were killed and healing was evaluated with mechanical, histological, and microcomputed tomography methods. Constant fixation stiffness control groups with either stiff or flexible fixation were included for comparison. Results - The callus stiffness of the stiff group and all 4 experimental groups was greater than in the flexible group. The callus of the flexible group was larger but contained a higher proportion of unmineralized tissue and cartilage. The stiff and modulated groups (3, 7, 14, and 21 days) all showed bony bridging at 5 weeks, as well as signs of callus remodeling. Stiffening fixation at 7 and 14 days after osteotomy produced the highest degree of callus bridging. Bone mineral density in the fracture gap was highest in animals in which the fixation was stiffened after 14 days. Interpretation - The predicted benefit of a large robust callus formed through early flexible fixation could not be shown, but the benefits of stabilizing a flexible construct to achieve timely healing were demonstrated at all time points.


Subject(s)
Bony Callus/physiopathology , External Fixators , Femoral Fractures/surgery , Femur/physiopathology , Fracture Fixation/methods , Fracture Healing , Animals , Biomechanical Phenomena , Bony Callus/diagnostic imaging , Bony Callus/pathology , Diaphyses , Femur/diagnostic imaging , Femur/pathology , Femur/surgery , Male , Osteotomy/methods , Random Allocation , Rats , Rats, Wistar , X-Ray Microtomography
3.
PLoS One ; 11(2): e0147465, 2016.
Article in English | MEDLINE | ID: mdl-26849055

ABSTRACT

Following bone fracture, the repair process starts with an inflammatory reaction at the fracture site. Fracture healing is disturbed when the initial inflammation is increased or prolonged, whereby, a balanced inflammatory response is anticipated to be crucial for fracture healing, because it may induce down-stream responses leading to tissue repair. However, the impact of the immune response on fracture healing remains poorly understood. Here, we investigated bone healing in NOD/scid-IL2Rγcnull mice, which exhibit severe defects in innate and adaptive immunity, by biomechanical testing, histomorphometry and micro-computed tomography. We demonstrated that NOD/scid-IL2Rγcnull mice exhibited normal skeletal anatomy and a mild bone phenotype with a slightly reduced bone mass in the trabecular compartment in comparison to immunocompetent Balb/c mice. Fracture healing was impaired in immunodeficient NOD/scid-IL2Rγcnull mice. Callus bone content was unaffected during the early healing stage, whereas it was significantly reduced during the later healing period. Concomitantly, the amount of cartilage was significantly increased, indicating delayed endochondral ossification, most likely due to the decreased osteoclast activity observed in cells isolated from NOD/scid-IL2Rγcnull mice. Our results suggest that--under aseptic, uncomplicated conditions--the immediate immune response after fracture is non-essential for the initiation of bone formation. However, an intact immune system in general is important for successful bone healing, because endochondral ossification is delayed in immunodeficient NOD/scid-IL2Rγcnull mice.


Subject(s)
Fracture Healing/genetics , Fracture Healing/immunology , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Animals , Biomechanical Phenomena , Bone Resorption/genetics , Bone Resorption/immunology , Disease Models, Animal , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , Osteogenesis/immunology , Phenotype , X-Ray Microtomography
4.
PLoS One ; 10(7): e0131194, 2015.
Article in English | MEDLINE | ID: mdl-26147725

ABSTRACT

In polytrauma patients a thoracic trauma is one of the most critical injuries and an important trigger of post-traumatic inflammation. About 50% of patients with thoracic trauma are additionally affected by bone fractures. The risk for fracture malunion is considerably increased in such patients, the pathomechanisms being poorly understood. Thoracic trauma causes regional alveolar hypoxia and, subsequently, hypoxemia, which in turn triggers local and systemic inflammation. Therefore, we aimed to unravel the role of oxygen in impaired bone regeneration after thoracic trauma. We hypothesized that short-term breathing of 100% oxygen in the early post-traumatic phase ameliorates inflammation and improves bone regeneration. Mice underwent a femur osteotomy alone or combined with blunt chest trauma 100% oxygen was administered immediately after trauma for two separate 3 hour intervals. Arterial blood gas tensions, microcirculatory perfusion and oxygenation were assessed at 3, 9 and 24 hours after injury. Inflammatory cytokines and markers of oxidative/nitrosative stress were measured in plasma, lung and fracture hematoma. Bone healing was assessed on day 7, 14 and 21. Thoracic trauma induced pulmonary and systemic inflammation and impaired bone healing. Short-term exposure to 100% oxygen in the acute post-traumatic phase significantly attenuated systemic and local inflammatory responses and improved fracture healing without provoking toxic side effects, suggesting that hyperoxia could induce anti-inflammatory and pro-regenerative effects after severe injury. These results suggest that breathing of 100% oxygen in the acute post-traumatic phase might reduce the risk of poorly healing fractures in severely injured patients.


Subject(s)
Acute Lung Injury/therapy , Femoral Fractures/therapy , Fracture Healing/drug effects , Oxygen Inhalation Therapy , Thoracic Injuries/therapy , Wounds, Nonpenetrating/therapy , Acute Lung Injury/etiology , Animals , Bone Regeneration/drug effects , Bony Callus/metabolism , Carbon Dioxide/blood , Cytokines/analysis , DNA Damage , Femoral Fractures/etiology , Hematoma/metabolism , Hyperoxia/metabolism , Hyperoxia/physiopathology , Inflammation Mediators/analysis , Lung/chemistry , Male , Mice , Mice, Inbred C57BL , Microcirculation , Osteotomy/adverse effects , Oxidative Stress , Oxygen/blood , Oxyhemoglobins/analysis , Random Allocation , Thoracic Injuries/classification , Thoracic Injuries/complications , Weight-Bearing , Wounds, Nonpenetrating/classification , Wounds, Nonpenetrating/complications
5.
Dis Model Mech ; 8(1): 93-104, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25381012

ABSTRACT

Fracture healing is impaired in aged and osteoporotic individuals. Because adequate mechanical stimuli are able to increase bone formation, one therapeutical approach to treat poorly healing fractures could be the application of whole-body vibration, including low-magnitude high-frequency vibration (LMHFV). We investigated the effects of LMHFV on fracture healing in aged osteoporotic mice. Female C57BL/6NCrl mice (n=96) were either ovariectomised (OVX) or sham operated (non-OVX) at age 41 weeks. When aged to 49 weeks, all mice received a femur osteotomy that was stabilised using an external fixator. The mice received whole-body vibrations (20 minutes/day) with 0.3 G: peak-to-peak acceleration and a frequency of 45 Hz. After 10 and 21 days, the osteotomised femurs and intact bones (contra-lateral femurs, lumbar spine) were evaluated using bending-testing, micro-computed tomography (µCT), histology and gene expression analyses. LMHFV disturbed fracture healing in aged non-OVX mice, with significantly reduced flexural rigidity (-81%) and bone formation (-80%) in the callus. Gene expression analyses demonstrated increased oestrogen receptor ß (ERß, encoded by Esr2) and Sost expression in the callus of the vibrated animals, but decreased ß-catenin, suggesting that ERß might mediate these negative effects through inhibition of osteoanabolic Wnt/ß-catenin signalling. In contrast, in OVX mice, LMHFV significantly improved callus properties, with increased flexural rigidity (+1398%) and bone formation (+637%), which could be abolished by subcutaneous oestrogen application (0.025 mg oestrogen administered in a 90-day-release pellet). On a molecular level, we found an upregulation of ERα in the callus of the vibrated OVX mice, whereas ERß was unaffected, indicating that ERα might mediate the osteoanabolic response. Our results indicate a major role for oestrogen in the mechanostimulation of fracture healing and imply that LMHFV might only be safe and effective in confined target populations.


Subject(s)
Estrogens/physiology , Fracture Healing , Receptors, Estrogen/blood , Vibration , Animals , Female , Femur/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Protein Biosynthesis , Signal Transduction , beta Catenin/metabolism
6.
J Orthop Res ; 32(12): 1589-95, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25183200

ABSTRACT

The healing process consists of at least three phases: inflammatory, repair, and remodeling phase. Because callus stiffness correlates with the healing phases, it is suitable for evaluating the fracture healing process. Our aim was to develop a method which allows determination of callus stiffness in vivo, the healing time and the duration of the repair phase. The right femurs of 16 Wistar rats were osteotomized and stabilized with either more rigid or more flexible external fixation. Fixator deformation was measured using strain gauges during gait analysis. The strains were recalculated as the callus stiffness over the time course of healing, and the healing phases were identified based on stiffness thresholds. Our hypothesis was that stabilization with more flexible external fixation prolongs the repair phase, therefore resulting in an extended healing time. Confirming our hypothesis, the duration of the repair phase (rigid: approximately 15 days, flexible: approximately 41 days) and the healing time (rigid: approximately 27 days, flexible: approximately 62 days) were significantly longer for more flexible external fixation. Our method allows the quantitative detection of differences in the healing time and duration of the repair phase without multiple time-point sacrifices, which reduces the number of animals in experimental studies.


Subject(s)
Bony Callus/physiology , Fracture Healing/physiology , Animals , Biomechanical Phenomena , Male , Rats, Wistar , Time Factors
7.
J Orthop Res ; 32(8): 1006-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24729351

ABSTRACT

Low-magnitude high-frequency vibration (LMHFV) provokes anabolic effects in non-fractured bone; however, in fracture healing, inconsistent results were reported and optimum vibration conditions remain unidentified. Here, we investigated frequency dependent effects of LMHFV on fracture healing. Twelve-week-old, female C57BL/6 mice received a femur osteotomy stabilized using an external fixator. The mice received whole-body vibrations (20 min/day) with 0.3g peak-to-peak acceleration and a frequency of either 35 or 45 Hz. After 10 and 21 days, the osteotomized femurs and intact bones (contra-lateral femurs, lumbar spine) were evaluated using bending-testing, µ-computed tomography, and histomorphometry. In non-fractured trabecular bone, vibration with 35 Hz significantly increased the relative amount of bone (+28%) and the trabecular number (+29%), whereas cortical bone was not influenced. LMHFV with 45 Hz failed to provoke anabolic effects in trabecular or cortical bone. Fracture healing was not significantly influenced by whole-body vibration with 35 Hz, whereas 45 Hz significantly reduced bone formation (-64%) and flexural rigidity (-34%) of the callus. Although the exact mechanisms remain open, our results suggest that small vibration setting changes could considerably influence LMHFV effects on bone formation in remodeling and repair, and even disrupt fracture healing, implicating caution when treating patients with impaired fracture healing.


Subject(s)
Fracture Healing/physiology , Vibration , Animals , Biomechanical Phenomena , Female , Mice , Mice, Inbred C57BL , Osteogenesis , X-Ray Microtomography
8.
Eur Spine J ; 23(6): 1361-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24566944

ABSTRACT

PURPOSE: Balloon kyphoplasty (BKP) with calcium phosphate cement (CPC) is increasingly being used for spinal surgery in younger patients. In routinely performed follow-up CT scans we observed considerable areas of demineralization in CPC processed vertebrae in several patients. To rule out infections or inflammations histological examinations were planned for these patients. METHODS: Ten patients (23-54 years; six men) with significant demineralization areas in CT scans after CPC balloon kyphoplasty were selected. Punch biopsies from these areas were taken in local anesthesia using a biopsy needle. One half of the specimen was decalcified and embedded in paraffin, and sections were examined histologically using hematoxylin and eosin, Van Gieson, and trichrome staining. The second half of the specimen was cast directly in methyl methacrylate and sections were examined by Paragon and von Kossa/Safranin staining. Stained slides were viewed under light microscopy. RESULTS: Bone-punch specimens were taken at 17.5 months (mean) after BKP with CPC. In most cases, the cement was well surrounded by newly formed lamellar bone with very tight connections between the cement and new bone. Unmineralized areas were observed sporadically at the cement surface and adjacent to the implant. There were no pronounced signs of inflammation or osteolysis of adjacent bone. No complications were observed during or following patients' biopsy procedures. CONCLUSIONS: CPC demonstrated good biocompatibility and osseointegration in clinical use, with no evidence of inflammation or osteonecrosis. Demineralized areas in CT scans could be a result of remodeling of the cancellous bone in vertebral bodies.


Subject(s)
Bone Cements , Bone Demineralization, Pathologic , Calcium Phosphates , Kyphoplasty , Spine/pathology , Adult , Biopsy, Needle , Humans , Male , Microscopy , Middle Aged , Osseointegration , Osteoblasts/pathology , Spinal Fractures/surgery , Tomography, X-Ray Computed , Young Adult
9.
Clin Oral Implants Res ; 25(11): 1239-1244, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24102843

ABSTRACT

PURPOSE: It is supposed that the demographic change will lead to an increase in patients with impaired alveolar bone conditions. Large animal models are of particular interest in this context as they are suitable for developing and testing new dental implants. Recently, we demonstrated that surgical hypothalamo-pituitary disconnection (HPD) causes a pronounced low-turnover situation leading to cortical and trabecular bone loss in sheep. In this study, we aimed to investigate the influence of the HPD procedure on the alveolar bone. METHODS: Ten adult Merino ewes were randomly assigned to two groups: Control and HPD. After 6 months, we analysed the cortical and trabecular bone of all mandibles by histomorphometry and high-resolution peripheral quantitative computed tomography (HR-pQCT). RESULTS: HPD ewes showed a significant decrease in cortical thickness by ~20%, a significant increase in cortical porosity by ~20% and a significant decrease in bone volume by ~30% in comparison with Control ewes. CONCLUSION: Our results underline the importance of central regulatory mechanisms of bone turnover. However, further studies are needed to understand these central regulatory elements of bone turnover in detail and to judge the value of the HPD sheep for dental research.


Subject(s)
Alveolar Bone Loss/etiology , Hypothalamic Diseases/complications , Hypothalamo-Hypophyseal System/physiopathology , Mandibular Diseases/etiology , Alveolar Bone Loss/diagnostic imaging , Alveolar Bone Loss/pathology , Animals , Bone Density/physiology , Bone Remodeling/physiology , Disease Models, Animal , Female , Mandibular Diseases/diagnostic imaging , Mandibular Diseases/pathology , Ovariectomy/methods , Random Allocation , Sheep , Time Factors , Tomography, X-Ray Computed/methods , X-Ray Microtomography/methods
10.
PLoS One ; 8(11): e81341, 2013.
Article in English | MEDLINE | ID: mdl-24260573

ABSTRACT

There is increasing evidence that complement may play a role in bone development. Our previous studies demonstrated that the key complement receptor C5aR was strongly expressed in the fracture callus not only by immune cells but also by bone cells and chondroblasts, indicating a function in bone repair. To further elucidate the role of complement in bone healing, this study investigated fracture healing in mice in the absence of the key complement molecules C3 and C5. C3(-/-) and C5(-/-) as well as the corresponding wildtype mice received a standardized femur osteotomy, which was stabilized using an external fixator. Fracture healing was investigated after 7 and 21 days using histological, micro-computed tomography and biomechanical measurements. In the early phase of fracture healing, reduced callus area (C3(-/-): -25%, p=0.02; C5(-/-): -20% p=0.052) and newly formed bone (C3(-/-): -38%, p=0.01; C5(-/-): -52%, p=0.009) was found in both C3- and C5-deficient mice. After 21 days, healing was successful in the absence of C3, whereas in C5-deficient mice fracture repair was significantly reduced, which was confirmed by a reduced bending stiffness (-45%; p=0.029) and a smaller callus volume (-17%; p=0.039). We further demonstrated that C5a was activated in C3(-/-) mice, suggesting cleavage via extrinsic pathways. Our results suggest that the activation of the terminal complement cascade in particular may be crucial for successful fracture healing.


Subject(s)
Bony Callus/immunology , Complement C3a/genetics , Complement C5a/genetics , Fracture Healing/genetics , Fractures, Bone/genetics , Animals , Biomechanical Phenomena , Bony Callus/diagnostic imaging , Bony Callus/pathology , Complement Activation/genetics , Complement C3a/deficiency , Complement C3a/immunology , Complement C5a/deficiency , Complement C5a/immunology , Elasticity , Femur/diagnostic imaging , Femur/injuries , Fracture Healing/immunology , Fractures, Bone/diagnostic imaging , Fractures, Bone/immunology , Gene Deletion , Gene Expression/immunology , Hardness , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Male , Mice , X-Ray Microtomography
11.
J Orthop Res ; 31(11): 1845-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23818033

ABSTRACT

Sphingosine-1-phosphate (S1P) has recently been recognized as a crucial coupling molecule of osteoclast and osteoblast activity provoking osteoanabolic effects. Targeting S1P receptors could, therefore, be a potential strategy to support bone formation in osteopenic diseases or in fracture repair. Here we investigated whether systemic treatment with the S1P analog FTY720 (Fingolimod) could improve fracture healing. Twelve-week-old, female C57BL/6 mice received an osteotomy of the femur, which was stabilized using an external fixator. The mice received a daily subcutaneous injection of either FTY720 (6 mg/kg) or vehicle from the third postoperative day. Fracture healing was evaluated after 10 and 21 days using biomechanical testing, µ-computed tomography, and histomorphometry. Because FTY720 is supposed to influence osteoclast recruitment, osteoclasts were identified in the fracture callus by staining for tartrate resistant acid phosphatase (TRAP). There were no significant differences in callus mechanical properties, tissue composition and osteoclast number between the groups, suggesting that systemically applied FTY720 did not influence bone regeneration in this model of regular fracture healing. Even if further studies should test the potency of FTY720 under unfavorable healing conditions, we conclude that the effect of systemically applied FTY720 on fracture healing might be inferior compared to other anabolic treatments. © 2013 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 31:1845-1850, 2013.


Subject(s)
Fracture Healing/drug effects , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Acid Phosphatase/metabolism , Animals , Biomechanical Phenomena , Bone Regeneration/drug effects , Female , Fingolimod Hydrochloride , Mice , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteoclasts/physiology , Sphingosine/pharmacology
12.
J Orthop Res ; 31(11): 1851-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23813786

ABSTRACT

We recently established a large animal model of osteoporosis in sheep using hypothalamic-pituitary disconnection (HPD). As central regulation is important for bone metabolism, HPD-sheep develop severe osteoporosis because of low bone turnover. In this study we investigated metaphyseal fracture healing in HPD-sheep. To elucidate potential pathomechanisms, we included a treatment group receiving thyroxine T4 and 17ß-estradiol. Because clinically osteoporotic fractures often occur in the bone metaphysis, HPD-sheep and healthy controls received an osteotomy in the distal femoral condyle. Half of the HPD-sheep were systemically treated with thyroxine T4 and 17ß-estradiol during the healing period. Fracture healing was evaluated after 8 weeks using pQCT, µCT, and histomorphometrical analysis. Bone mineral density (BMD) and bone volume/total volume (BV/TV) were considerably reduced by 30% and 36%, respectively, in the osteotomy gap of the HPD-sheep compared to healthy sheep. Histomorphometry also revealed a decreased amount of newly formed bone (-29%) and some remaining cartilage in the HPD-group, suggesting that HPD disturbed fracture healing. Thyroxine T4 and 17ß-estradiol substitution considerably improved bone healing in the HPD-sheep. Our results indicate that fracture healing requires central regulation and that thyroxine T4 and 17ß-estradiol contribute to the complex pathomechanisms of delayed metaphyseal bone healing in HPD-sheep.


Subject(s)
Fracture Healing , Hypothalamus/physiology , Osteoporotic Fractures/physiopathology , Pituitary Gland/physiology , Animals , Disease Models, Animal , Estradiol/therapeutic use , Fracture Healing/drug effects , Osteoporotic Fractures/drug therapy , Sheep , Thyroxine/therapeutic use
13.
J Trauma Acute Care Surg ; 74(2): 531-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23354247

ABSTRACT

BACKGROUND: We recently demonstrated that a blunt chest trauma, a strong inducer of the posttraumatic systemic inflammatory response and one of the most critical injuries in polytrauma patients, significantly delayed fracture healing in rats, possibly by the interaction of the systemic inflammation with early regeneration processes locally at the fracture site. The underlying cellular mechanisms, however, have as yet remained unknown. Therefore, the aim of this study was to analyze the cellular and morphologic composition of the early fracture callus after a blunt chest trauma. METHODS: Rats received an osteotomy of the right femur stabilized by an external fixator in combination with a blunt chest trauma or not. The animals were killed after 3, 7, and 35 days, and the fracture calli were analyzed histologically for new tissue formation, polymorphonuclear leucocytes, macrophages, osteoclasts, and the presence of the proinflammatory cytokine interleukin 6. RESULTS: The blunt chest trauma considerably increased the number of polymorphonuclear leucocytes in the callus by Day 3 compared with animals with isolated fractures. The number of macrophages was significantly reduced by the thoracic trauma at Days 3 and 7. The number of osteoclasts was not changed at any postoperative time point. After 3 days, the blunt chest trauma led to a significantly stronger interleukin 6 staining within the periosteal callus in zones of intramembranous ossification. During the time of cortical bridging at Day 35, the amount of newly formed bone was significantly decreased after blunt chest trauma. CONCLUSION: Our results suggest that the systemic posttraumatic inflammation induced by a thoracic trauma disturbed the inflammatory balance during the early healing stage by altering the recruitment of inflammatory cells and cytokine expression locally at the fracture site and thus impaired fracture healing. These findings provide new insights in the pathomechanisms of impaired fracture healing in patients experiencing severe trauma.


Subject(s)
Bony Callus/physiopathology , Femoral Fractures/physiopathology , Systemic Inflammatory Response Syndrome/etiology , Thoracic Injuries/complications , Animals , Bony Callus/chemistry , Bony Callus/cytology , Bony Callus/pathology , Disease Models, Animal , Interleukin-6/analysis , Interleukin-6/physiology , Macrophages/physiology , Male , Multiple Trauma/physiopathology , Neutrophils/physiology , Osteoclasts/physiology , Rats , Rats, Wistar , Systemic Inflammatory Response Syndrome/physiopathology , Time Factors
14.
J Orthop Res ; 31(3): 465-71, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23070742

ABSTRACT

In poly-traumatic patients, second hits are known to potentiate the posttraumatic systemic inflammatory response, thus increasing the risk of multi-organ dysfunction. In accordance with "damage control orthopaedic surgery" principles, fractures are initially treated with external fixators, which are replaced by internal osteosynthesis once the immunological status of the patient is considered stable. Recently, we demonstrated that a severe trauma impaired the healing of fractures stabilized by external fixation during the entire healing period. The question arose, whether switching to intramedullary nailing increases the inflammatory response in terms of a second hit, leading to a further impairment of bone healing. Wistar rats received a femoral osteotomy stabilized by an external fixator. Simultaneously half of the rats underwent an additional thoracic trauma. After 4 days, the external fixator was replaced by an intramedullary nail in half of the rats of the two groups. The inflammatory response was evaluated by measuring serum C5a levels. Fracture healing was determined by three-point-bending, µCT, and histomorphometry. The thoracic trauma significantly increased C5a concentrations 6, 24, and 72 h after the second surgical intervention. After 40 days, conversion to intramedullary nailing considerably decreased the flexural rigidity of the callus, with no significant differences between rats with or without thoracic trauma. After 47 days, flexural rigidity in rats subjected to conversion remained decreased compared to animals solely treated by external fixation, particularly in combination with blunt chest trauma. The results indicate that accumulation of second hits after multiple injuries could lead to aggravation of the fracture healing outcome.


Subject(s)
Bone Nails , Conversion to Open Surgery/adverse effects , External Fixators , Femoral Fractures/surgery , Fracture Healing/physiology , Animals , Biomechanical Phenomena/physiology , Complement C5a/metabolism , Disease Models, Animal , Femoral Fractures/diagnostic imaging , Femoral Fractures/physiopathology , Male , Osteotomy/methods , Rats , Rats, Wistar , Trauma Severity Indices , Wounds, Nonpenetrating/diagnostic imaging , Wounds, Nonpenetrating/physiopathology , Wounds, Nonpenetrating/surgery , X-Ray Microtomography
15.
PLoS One ; 8(12): e84232, 2013.
Article in English | MEDLINE | ID: mdl-24391920

ABSTRACT

Wnt signaling is a key regulator of bone metabolism and fracture healing. The canonical Wnt/ß-catenin pathway is regarded as the dominant mechanism, and targeting this pathway has emerged as a promising strategy for the treatment of osteoporosis and poorly healing fractures. In contrast, little is known about the role of non-canonical Wnt signaling in bone. Recently, it was demonstrated that the serpentine receptor Fzd9, a Wnt receptor of the Frizzled family, is essential for osteoblast function and positively regulates bone remodeling via the non-canonical Wnt pathway without involving ß-catenin-dependent signaling. Here we investigated whether the Fzd9 receptor is essential for fracture healing using a femur osteotomy model in Fzd9(-/-) mice. After 10, 24 and 32 days the fracture calli were analyzed using biomechanical testing, histomorphometry, immunohistochemistry, and micro-computed tomography. Our results demonstrated significantly reduced amounts of newly formed bone at all investigated healing time points in the absence of Fzd9 and, accordingly, a decreased mechanical competence of the callus tissue in the late phase of fracture healing. In contrast, cartilage formation and numbers of osteoclasts degrading mineralized matrix were unaltered. ß-Catenin immunolocalization showed that canonical Wnt-signaling was not affected in the absence of Fzd9 in osteoblasts as well as in proliferating and mature chondrocytes within the fracture callus. The expression of established differentiation markers was not altered in the absence of Fzd9, whereas chemokines Ccl2 and Cxcl5 seemed to be reduced. Collectively, our results suggest that non-canonical signaling via the Fzd9 receptor positively regulates intramembranous and endochondral bone formation during fracture healing, whereas it does not participate in the formation of cartilage or in the osteoclastic degradation of mineralized matrix. The finding that Fzd9, in addition to its role in physiological bone remodeling, regulates bone repair may have implications for the development of treatments for poorly or non-healing fractures.


Subject(s)
Fracture Healing/physiology , Osteogenesis/physiology , Receptors, Neurotransmitter/metabolism , Wnt Signaling Pathway/physiology , Animals , Biomechanical Phenomena , Bony Callus/physiology , Frizzled Receptors , Histological Techniques , Mice , Statistics, Nonparametric , X-Ray Microtomography
16.
J Orthop Res ; 30(4): 581-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21922535

ABSTRACT

Confirming clinical evidence, we recently demonstrated that a blunt chest trauma considerably impaired fracture healing in rats, possibly via the interaction of posttraumatic systemic inflammation with local healing processes, the underlying mechanisms being unknown. An important trigger of systemic inflammation is the complement system, with the potent anaphylatoxin C5a. Therefore, we investigated whether the impairment of fracture healing by a severe trauma resulted from systemically activated complement. Rats received a blunt chest trauma and a femur osteotomy stabilized with an external fixator. To inhibit the C5a-dependent posttraumatic systemic inflammation, half of the rats received a C5aR-antagonist intravenously immediately and 12 h after the thoracic trauma. Compared to the controls (control peptide), the treatment with the C5aR-antagonist led to a significantly increased flexural rigidity (three-point-bending test), an improved bony bridging of the fracture gap, and a slightly larger and qualitatively improved callus (µCT, histomorphometry) after 35 days. In conclusion, immunomodulation by a C5aR-antagonist could abolish the deleterious effects of a thoracic trauma on fracture healing, possibly by influencing the function of inflammatory and bone cells locally at the fracture site. C5a could possibly represent a target to prevent delayed bone healing in patients with severe trauma.


Subject(s)
Femoral Fractures/immunology , Fracture Healing/drug effects , Peptides, Cyclic/pharmacology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Thoracic Injuries/immunology , Wounds, Nonpenetrating/immunology , Animals , Biomechanical Phenomena/physiology , Complement C5a/antagonists & inhibitors , Complement C5a/immunology , Disease Models, Animal , Femoral Fractures/diagnostic imaging , Femoral Fractures/physiopathology , Fracture Healing/immunology , Inflammation/complications , Inflammation/drug therapy , Inflammation/physiopathology , Male , Osteotomy , Rats , Rats, Wistar , Receptor, Anaphylatoxin C5a/immunology , Thoracic Injuries/physiopathology , Trauma Severity Indices , Wounds, Nonpenetrating/physiopathology , X-Ray Microtomography
17.
J Orthop Res ; 29(5): 734-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21437953

ABSTRACT

In poly-traumatic patients a blunt chest trauma is an important trigger of the posttraumatic systemic inflammatory response. There is clinical evidence that fracture healing is delayed in such patients, however, experimental data are lacking. Therefore, we investigated the influence of a thoracic trauma on fracture healing in a rat model. Male Wistar rats received either a blunt chest trauma combined with a femur osteotomy or an isolated osteotomy. A more rigid or a more flexible external fixator was used for fracture stabilization to analyze whether the thoracic trauma influences regular healing and mechanically induced delayed bone healing differently. The blunt chest trauma induced a significant increase of IL-6 serum levels after 6 and 24 h, suggesting the induction of a systemic inflammation, whereas the isolated fracture had no effect. Under a more rigid fixation the thoracic trauma considerably impaired fracture healing after 35 days, reflected by a significantly reduced flexural rigidity (three-point-bending test), as well as a significantly diminished callus volume, moment of inertia, and relative bone surface (µCT analysis). In confirming the clinical evidence, this study reports for the first time that a blunt chest trauma considerably impaired bone healing, possibly via the interaction of the induced systemic inflammation with local inflammatory processes.


Subject(s)
Femoral Fractures/therapy , Fracture Healing/physiology , Thoracic Injuries/physiopathology , Wounds, Nonpenetrating/physiopathology , Animals , Biomechanical Phenomena , External Fixators , Femoral Fractures/surgery , Inflammation/etiology , Interleukin-6/blood , Male , Rats , Rats, Wistar , Thoracic Injuries/complications , Wounds, Nonpenetrating/complications
18.
Bone ; 48(4): 945-51, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21185956

ABSTRACT

The adaptive response of bone to load is dependent on molecular factors, including growth factor signaling, which is involved in the regulation of proliferation, differentiation and function of osteoblasts and osteoclasts. Based on a recent study, which has shown that the deficiency of growth factor midkine (Mdk) in mice at 12 and 18 months of age resulted in increased trabecular bone formation, we hypothesized that mechanically-induced bone remodeling may, at least in part, be dependent on Mdk expression. To investigate this, we loaded the ulnae of Mdk-deficient mice and appropriate wild-type mice at the age of 12 months using the in vivo ulna loading model. Histomorphometric quantification of the periosteal bone demonstrated an increased mineralizing surface, mineral apposition rate, and bone formation rate in ulnae of Mdk-deficient mice compared to wild-type mice in response to loading. Because Mdk has been shown to bind to a complex of receptor-type protein tyrosine phosphatase zeta (Ptprz) and low density lipoprotein receptor-related protein-6 (Lrp-6) together with the α4ß1- and α6ß1-integrins, we performed in vitro studies using osteoblastic cells, transiently over-expressing Mdk, Wnt-3a, and Ptprz to evaluate whether Mdk has a role in regulating bone formation by modulating Wnt signaling. We observed a negative effect of Mdk on Wnt signaling, the extent of which appeared to be dependent on Ptprz expression. Moreover, we performed in vitro loading studies with osteoblasts treated with recombinant Mdk and observed a negative effect on the expression of Wnt target genes, which play a critical role in osteoblast proliferation. In summary, our data demonstrate that Mdk-deficiency in mice has an anabolic effect on mechanically induced cortical bone formation. This could be due to an improved osteoblast function based on an enhancement of ß-catenin-dependent Wnt signaling by both Mdk-deficiency and mechanical loading.


Subject(s)
Bone and Bones/physiopathology , Cytokines/genetics , Stress, Physiological , Animals , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , Mice , Mice, Inbred C57BL , Midkine , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...