Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.080
Filter
Add more filters

Publication year range
1.
Hum Genomics ; 18(1): 87, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39148098

ABSTRACT

BACKGROUND: Recent studies suggested that genetic variants associated with monogenic bone disorders were involved in the pathogenesis of atypical femoral fractures (AFF). Here, we aim to identify rare genetic variants by whole exome sequencing in genes involved in monogenic rare skeletal diseases in 12 women with AFF and 4 controls without any fracture. RESULTS: Out of 33 genetic variants identified in women with AFF, eleven (33.3%) were found in genes belonging to the Wnt pathway (LRP5, LRP6, DAAM2, WNT1, and WNT3A). One of them was rated as pathogenic (p.Pro582His in DAAM2), while all others were rated as variants of uncertain significance according to ClinVar and ACMG criteria. CONCLUSIONS: Osteoporosis, rare bone diseases, and AFFs may share the same genes, thus making it even more difficult to identify unique risk factors.


Subject(s)
Exome Sequencing , Femoral Fractures , Low Density Lipoprotein Receptor-Related Protein-5 , Low Density Lipoprotein Receptor-Related Protein-6 , Humans , Female , Femoral Fractures/genetics , Femoral Fractures/pathology , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Middle Aged , Aged , Genetic Predisposition to Disease , Wnt1 Protein/genetics , Wnt3A Protein/genetics , Wnt Signaling Pathway/genetics , Osteoporosis/genetics , Osteoporosis/pathology , Bone Diseases/genetics , Case-Control Studies
2.
Biochem Biophys Res Commun ; 703: 149634, 2024 04 09.
Article in English | MEDLINE | ID: mdl-38354465

ABSTRACT

Fractures are frequent and severe musculoskeletal injuries. This study aimed to investigate the function of tenascin-C (TNC) in regulating chondrogenic during fracture healing and elucidate the underlying molecular mechanisms. A well-established femur fracture model in male C57BL/6J mice was used to transect the middle diaphysis of the femur. To identify the essential role of TNC, shTNC lentiviruses or TNC protein were administered in the animal model. Micro-CT analysis, histologic analysis, immunostaining assays, and gene expression analysis were employed to investigate the effect of TNC during fracture healing. An in vitro mesenchymal stem cell culture system was developed to investigate the role and molecular mechanism of TNC in regulating chondrogenesis. TNC expression was induced at the inflammatory phase and peaked at the cartilaginous callus phase during fracture healing. Knockdown of TNC expression in callus results in decreased callus formation and impaired fracture healing. Conversely, administration of exogenous TNC promoted chondrogenic differentiation, cartilage template formation and ultimately improved fracture healing. Both the Hedgehog and Hippo signaling pathways were found to be involved in the pro-chondrogenic function of TNC. Our observations demonstrate that TNC is a crucial factor responsible for endochondral ossification in fracture healing and provide a potential therapeutic strategy for promoting fracture healing.


Subject(s)
Femoral Fractures , Fracture Healing , Osteogenesis , Tenascin , Animals , Male , Mice , Bony Callus/pathology , Femoral Fractures/pathology , Hedgehogs , Hippo Signaling Pathway , Mice, Inbred C57BL , Tenascin/genetics , Tenascin/metabolism
3.
Int J Exp Pathol ; 105(2): 52-63, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38152045

ABSTRACT

Bone fractures are the most common form of musculoskeletal trauma worldwide. Numerous microRNAs (miRNAs) have been suggested to be participants in regulating bone-related diseases. Recent studies revealed the regulatory role of miR-22-3p in osteogenic differentiation, but its role in fracture healing has not been investigated previously. Here, a rat femoral fracture model was established, Bone marrow mesenchymal stem cells (BMSCs) were isolated to detect the specific function and underlying mechanisms of miR-22-3p. MiR-22-3p and sclerostin domain-containing 1 (SOSTDC1) expression was determined by RT-qPCR and immunohistochemistry staining. The levels of proteins associated with osteogenic differentiation were assessed by western blotting. Flow cytometry was conducted to identify the isolated rat BMSCs. Alizarin red staining, alkaline phosphatase staining and Oil Red O staining were used to evaluate the osteogenic and adipogenic differentiation of rat BMSCs. The interaction between miR-22-3p and SOSTDC1 was verified using a luciferase reporter assay. Haematoxylin and Eosin (H&E) staining of the bone tissues was performed to analyse the effect of miR-22-3p on histopathological changes in vivo. MiR-22-3p was downregulated in the callus tissues of rat femoral fracture, while the expression of SOSTDC1 was upregulated. The isolated rat BMSCs had the capacity for both osteogenic and adipogenic differentiation. The differentiation capacity of BMSCs into osteoblasts was increased by miR-22-3p overexpression. MiR-22-3p activated the PI3K/AKT pathway by targeting SOSTDC1. SOSTDC1 overexpression and PI3K/AKT signalling inhibitor LY294002 abolished the enhancing effect of miR-22-3p overexpression on the osteogenesis of BMSCs. Thus MiR-22-3p facilitated the femoral fracture healing in rats. MiR-22-3p overexpression promoted fracture healing via the activation of PI3K/AKT pathway by targeting SOSTDC1.


Subject(s)
Femoral Fractures , Mesenchymal Stem Cells , MicroRNAs , Animals , Humans , Rats , Adaptor Proteins, Signal Transducing/genetics , Cell Differentiation , Cells, Cultured , Femoral Fractures/genetics , Femoral Fractures/metabolism , Femoral Fractures/pathology , Fracture Healing , MicroRNAs/genetics , MicroRNAs/metabolism , Osteogenesis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
4.
Int J Legal Med ; 138(4): 1685-1700, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38376570

ABSTRACT

When dealing with badly preserved cadavers or skeletal human remains, the assessment of death circumstances remains challenging. When forensic evidence cannot be taken from the skin and soft tissue, the information may only be deduced from more resistant elements such as bone. Compared to cranial gunshot injuries, reliable data on ballistic long bone trauma remains scarce. This study aims to define ballistic fracture characteristics in human long bones. The shaft of 16 femurs and 13 humeri from body donors was perpendicularly shot with a 9-mm Luger full metal jacket bullet at an impact velocity of 360 m/s from a distance of 2 m. Some bones were embedded in Clear Ballistics Gel®, and some were shot without soft tissue simulant in order to better visualise the fracture propagation on the high-speed camera. The fractures were examined macroscopically and compared between the sample groups. We consistently found comminuted fractures with a stellate pattern. Fracture details were classified into entrance, exit and general characteristics. For some traits, we detected different occurrence values in the group comparison. The results indicate that some of the traits depend on bone properties such as shaft diameter, bone length and cortical thickness. The presence of ballistic gel also influenced some fracture traits, emphasising the relevance of soft tissue simulant in osseous gunshot experiments. This study revealed new insights in the detailed fracture pattern of human long bones. These may serve as guidelines for the identification and reconstruction of gunshot trauma in human long bones.


Subject(s)
Forensic Ballistics , Wounds, Gunshot , Humans , Forensic Ballistics/methods , Wounds, Gunshot/pathology , Male , Middle Aged , Fractures, Comminuted/pathology , Adult , Aged , Female , Femoral Fractures/pathology
5.
J Appl Biomed ; 22(2): 67-73, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38912861

ABSTRACT

BACKGROUND AND OBJECTIVES: We aimed to determine the effects of vanillic acid (VA) on fracture healing radiologically, histologically, immunohistochemically, and biomechanically using a rat femur open fracture injury model. METHODS: 32 male Wistar-Albino rats were used and divided into two groups: the study group (VA) and the control group. From the time they were operated on until they were sacrificed, the rats in the study group were given 100 mg/kg/day VA by oral gavage. After sacrification, the femurs were analyzed. RESULTS: It was observed that the Huo histological scoring was significantly higher in the VA group (p = 0.001), and the ratio of the amount of callus tissue compared to intact bone tissue was significantly higher. While no significant difference was observed in immunohistochemical H-scores in ColI antibody staining (p = 1.000), a borderline significant difference in favor of VA was observed in ColIII antibody staining (p = 0.078). In biomechanical analysis, failure load (N), total energy (J), maximum stress (MPa), and stiffness (N/mm) measurements were significantly higher in the VA group (p = 0.040, p = 0.021, p = 0.015, and p = 0.035, respectively). CONCLUSION: It has been observed that VA, with its antioxidative properties, increases fracture healing in rats, in which an open fracture model was created. We are hopeful that such an antioxidant, which is common in nature, will increase fracture healing. Since this study is the first to examine the effect of VA on fracture healing, further studies are needed.


Subject(s)
Femoral Fractures , Fracture Healing , Rats, Wistar , Vanillic Acid , Animals , Vanillic Acid/pharmacology , Vanillic Acid/therapeutic use , Fracture Healing/drug effects , Male , Femoral Fractures/drug therapy , Femoral Fractures/pathology , Rats , Disease Models, Animal , Biomechanical Phenomena/drug effects , Femur/drug effects , Femur/pathology , Bony Callus/drug effects , Bony Callus/pathology
6.
BMC Musculoskelet Disord ; 24(1): 65, 2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36694156

ABSTRACT

BACKGROUND: Correctly identifying patients at risk of femoral fracture due to metastatic bone disease remains a clinical challenge. Mirels criteria remains the most widely referenced method with the advantage of being easily calculated but it suffers from poor specificity. The purpose of this study was to develop and evaluate a modified Mirels scoring system through scoring modification of the original Mirels location component within the proximal femur. METHODS: Computational (finite element) experiments were performed to quantify strength reduction in the proximal femur caused by simulated lytic lesions at defined locations. Virtual spherical defects representing lytic lesions were placed at 32 defined locations based on axial (4 axial positions: neck, intertrochanteric, subtrochanteric or diaphyseal) and circumferential (8 circumferential: 45-degree intervals) positions. Finite element meshes were created, material property assignment was based on CT mineral density, and femoral head/greater trochanter loading consistent with stair ascent was applied. The strength of each femur with a simulated lesion divided by the strength of the intact femur was used to calculate the Location-Based Strength Fraction (LBSF). A modified Mirels location score was next defined for each of the 32 lesion locations with an assignment of 1 (LBSF > 75%), 2 (LBSF: 51-75%), and 3 (LBSF: 0-50%). To test the new scoring system, data from 48 patients with metastatic disease to the femur, previously enrolled in a Musculoskeletal Tumor Society (MSTS) cross-sectional study was used. The lesion location was identified for each case based on axial and circumferential location from the CT images and assigned an original (2 or 3) and modified (1,2, or 3) Mirels location score. The total score for each was then calculated. Eight patients had a fracture of the femur and 40 did not over a 4-month follow-up period. Logistic regression and decision curve analysis were used to explore relationships between clinical outcome (Fracture/No Fracture) and the two Mirels scoring methods. RESULTS: The location-based strength fraction (LBSF) was lowest for lesions in the subtrochanteric and diaphyseal regions on the lateral side of the femur; lesions in these regions would be at greatest risk of fracture. Neck lesions located at the anterior and antero-medial positions were at the lowest risk of fracture. When grouped, neck lesions had the highest LBSF (83%), followed by intertrochanteric (72%), with subtrochanteric (50%) and diaphyseal lesions (49%) having the lowest LBSF. There was a significant difference (p < 0.0001) in LBSF between each axial location, except subtrochanteric and diaphyseal which were not different from each other (p = 0.96). The area under the receiver operator characteristic (ROC) curve using logistic regression was greatest for modified Mirels Score using site specific location of the lesion (Modified Mirels-ss, AUC = 0.950), followed by a modified Mirels Score using axial location of lesion (Modified Mirels-ax, AUC = 0.941). Both were an improvement over the original Mirels score (AUC = 0.853). Decision curve analysis was used to quantify the relative risks of identifying patients that would fracture (TP, true positives) and those erroneously predicted to fracture (FP, false positives) for the original and modified Mirels scoring systems. The net benefit of the scoring system weighed the benefits (TP) and harms (FP) on the same scale. At a threshold probability of fracture of 10%, use of the modified Mirels scoring reduced the number of false positives by 17-20% compared to Mirels scoring. CONCLUSIONS: A modified Mirels scoring system, informed by detailed analysis of the influence of lesion location, improved the ability to predict impending pathological fractures of the proximal femur for patients with metastatic bone disease. Decision curve analysis is a useful tool to weigh costs and benefits concerning fracture risk and could be combined with other patient/clinical factors that contribute to clinical decision making.


Subject(s)
Bone Diseases , Femoral Fractures , Neoplasms , Humans , Cross-Sectional Studies , Femur/diagnostic imaging , Femur/pathology , Femoral Fractures/diagnostic imaging , Femoral Fractures/etiology , Femoral Fractures/pathology , Bone Diseases/pathology , Finite Element Analysis
7.
Eur J Orthop Surg Traumatol ; 33(8): 3597-3601, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37246990

ABSTRACT

PURPOSE: Pathologic fractures of the extremities due to carcinoma metastases require individual and patient prognosis-related stabilization procedures. Quick remobilization of the patient to restore the quality of life is of high importance, especially in the case of subtrochanteric and diaphyseal femoral fractures. In our retrospective cohort study, we evaluated intraoperative blood loss, length of operation, complication rate, and regain of lower extremity function in plate compound osteosynthesis (PCO) versus intramedullary nailing (IM) for subtrochanteric and diaphyseal pathologic fractures of the femur. METHODS: Between January 2010 and July 2021, we retrospectively reviewed 49 patients who were treated at our institution for pathologic fractures of the subtrochanteric and diaphyseal femurs for group differences in terms of blood loss, length of operation, implant survival, and Musculoskeletal Tumor Society (MSTS) score. RESULTS: We included 49 stabilization procedures of the lower extremity due to pathologic fractures of the proximal or diaphyseal femur, with a mean follow-up of 17.7 months. IM (n = 29) had a significantly shorter operation time than PCO (n = 20) (112.4 ± 9.4 and 163.3 ± 15.96 min, respectively). We did not detect any significant differences in terms of blood loss, complication rate, implant survival, or MSTS score. CONCLUSION: Based on our data, pathologic subtrochanteric and diaphyseal fractures of the femur can be stabilized with IM, which has a shorter operation time than PCO, but the complication rate, implant survival, and blood loss remain unaffected.


Subject(s)
Femoral Fractures , Fracture Fixation, Intramedullary , Fractures, Spontaneous , Musculoskeletal Diseases , Humans , Fractures, Spontaneous/etiology , Fractures, Spontaneous/surgery , Fracture Fixation, Intramedullary/adverse effects , Fracture Fixation, Intramedullary/methods , Retrospective Studies , Quality of Life , Femoral Fractures/etiology , Femoral Fractures/surgery , Femoral Fractures/pathology , Lower Extremity , Treatment Outcome , Bone Nails/adverse effects
8.
Biochem Biophys Res Commun ; 634: 168-174, 2022 12 17.
Article in English | MEDLINE | ID: mdl-36244115

ABSTRACT

Classical histological methods such as hematoxylin-eosin staining, have been, and in some areas still are, an important benchmark for the evaluation of biological tissues. However, the current method of assessment is primarily a qualitative assessment of the tissue under investigation. The aim of this paper is to contribute to the improvement of classical histological methods, by applying physical techniques that allow objective, quantitative data to be added to qualitative assessments, especially in areas where conflicting results are available. To this end, the effect of hypolipidemic medication on the callus formation process of normal bone and pathological osteoporotic bone was investigated. The study allowed us to associate UV-VIS spectroscopy wave number with specific hematoxylin-eosin staining of different types of bone tissue structures, the evolving structures in the callus formation process. This association allowed the quantitative assessment of the callusing process in ovariectomized (associated with pathological, osteoporotic bone) and non-ovariectomized (associated with normal bone) rats, with three groups - the control group, simvastatin-treated group, and fenofibrate-treated group. The study showed that in the non-ovariectomized groups both treatments delayed callus formation. In the ovariectomized groups, simvastatin delayed and fenofibrate promoted callus formation.


Subject(s)
Femoral Fractures , Fenofibrate , Osteoporosis , Female , Humans , Rats , Animals , Rats, Wistar , Fracture Healing , Fenofibrate/pharmacology , Eosine Yellowish-(YS)/pharmacology , Eosine Yellowish-(YS)/therapeutic use , Hematoxylin/pharmacology , Hematoxylin/therapeutic use , Rats, Sprague-Dawley , Ovariectomy , Bony Callus/pathology , Femoral Fractures/diagnostic imaging , Femoral Fractures/pathology , Osteoporosis/drug therapy , Osteoporosis/pathology , Femur/pathology , Spectrum Analysis , Simvastatin/pharmacology , Simvastatin/therapeutic use
9.
J Bone Miner Metab ; 40(4): 704-711, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35637395

ABSTRACT

INTRODUCTION: Prostate cancer often forms osteoblastic lesions that appear as a high-dense shadow upon X-ray. Although the lesions may seem to increase bone strength, pathological fracture occurs in one in four patients with prostate cancer. The aim of this study is to elucidate the factors that may increase the risk of pathological fracture in patients with prostate cancer metastases in the proximal femur by analyzing computed tomography data. MATERIALS AND METHODS: Computed tomography data of the femur of 62 prostate cancer patients were retrospectively analyzed. The patients were divided into three groups based on the presence or absence of femoral metastatic lesions and pathological fracture. Surgical specimens of the proximal femur collected from patients who had a pathological fracture were histologically analyzed. RESULTS: Bone density in the marrow area was increased in all cases with metastases compared with those with no metastases. Contrarily, the cortical bone density at the medial trochanter region was significantly lower in patients who had pathological fractures in the proximal femur than those who did not. Accordingly, histological analysis of the surgical specimens revealed that the affected cortical bone was osteopenic without any apparent new bone formation. CONCLUSION: These results indicate that prostate cancer is less effective in inducing bone formation in the cortex than in the marrow and that the decrease in the cortical bone density at the medial trochanter region leads to an increased risk of pathological fracture. Therefore, a previously undocumented risk factor for pathological fracture in prostate cancer patients is presented.


Subject(s)
Femoral Fractures , Fractures, Spontaneous , Prostatic Neoplasms , Bone Density , Femoral Fractures/diagnostic imaging , Femoral Fractures/pathology , Femur/diagnostic imaging , Femur/pathology , Fractures, Spontaneous/complications , Fractures, Spontaneous/pathology , Humans , Male , Prostatic Neoplasms/complications , Retrospective Studies , Risk Assessment , Tomography, X-Ray Computed/methods
10.
Arch Orthop Trauma Surg ; 142(10): 2597-2609, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34097123

ABSTRACT

INTRODUCTION: Distal femur fractures are challenging injuries historically associated with high rates of nonunion and varus collapse with operative management. As a result, clinical and research interest in dual plating (DP) of distal femur fractures has seen a dramatic increase in recent years. The purpose of this study was to systematically review the literature regarding vascular anatomy and biomechanics of distal femur fractures treated with DP constructs. MATERIALS AND METHODS: A systematic literature review of two medical databases (PubMed & Scopus) was performed to identify peer-reviewed studies on the anatomy and biomechanics regarding DP of distal femur fractures. A total of 1,001 papers were evaluated and 14 papers met inclusion criteria (6 anatomy and 8 biomechanics). Methodological quality scores were used to assess quality and potential bias in the included studies. RESULTS: In the biomechanical studies, DP constructs demonstrated greater axial and rotational stiffness, as well as less displacement and fewer incidences of failure compared to all other constructs. Vascular studies showed that the femoral artery crosses the mid-shaft femur approximately 16.0-18.8 cm proximal to the adductor tubercle and it is located on average 16.6-31.1 mm from the femoral shaft at this location, suggesting that medial plate application can be achieved safely in the distal femur. The methodological quality of the included studies was good for biomechanical studies (Traa score 79.1; range 53-92.5) and excellent for anatomical studies (QUACs score 81.9; range 69.0-88.5). CONCLUSIONS: Existing biomechanics literature suggests that DP constructs are mechanically stronger than other constructs commonly used in the treatment of distal femur fractures. Furthermore, medial distal femoral anatomy allows for safe application of DP constructs, even in a minimally invasive fashion. Dual plating should be considered for patients with distal femur fractures that have risk factors for instability, varus collapse, or nonunion.


Subject(s)
Femoral Fractures , Fracture Fixation, Internal , Biomechanical Phenomena , Bone Plates , Femoral Fractures/pathology , Femoral Fractures/surgery , Fracture Fixation, Internal/methods , Humans
11.
FASEB J ; 34(4): 5673-5687, 2020 04.
Article in English | MEDLINE | ID: mdl-32115776

ABSTRACT

Surface chemistry and mechanical stability determine the osteogenic capability of bone implants. The development of high-strength bioactive scaffolds for in-situ repair of large bone defects is challenging because of the lack of satisfying biomaterials. In this study, highly bioactive Ca-silicate (CSi) bioceramic scaffolds were fabricated by additive manufacturing and then modified for pore-wall reinforcement. Pure CSi scaffolds were fabricated using a direct ink writing technique, and the pore-wall was modified with 0%, 6%, or 10% Mg-doped CSi slurry (CSi, CSi-Mg6, or CSi-Mg10) through electrostatic interaction. Modified CSi@CSi-Mg6 and CSi@CSi-Mg10 scaffolds with over 60% porosity demonstrated an appreciable compressive strength beyond 20 MPa, which was ~2-fold higher than that of pure CSi scaffolds. CSi-Mg6 and CSi-Mg10 coating layers were specifically favorable for retarding bio-dissolution and mechanical decay of scaffolds in vitro. In-vivo investigation of critical-size femoral bone defects repair revealed that CSi@CSi-Mg6 and CSi@CSi-Mg10 scaffolds displayed limited biodegradation, accelerated new bone ingrowth (4-12 weeks), and elicited a suitable mechanical response. In contrast, CSi scaffolds exhibited fast biodegradation and retarded new bone regeneration after 8 weeks. Thus, tailoring of the chemical composition of pore-wall struts of CSi scaffolds is beneficial for enhancing the biomechanical properties and bone repair efficacy.


Subject(s)
Biocompatible Materials/chemistry , Bone and Bones/cytology , Calcium Compounds/chemistry , Femoral Fractures/therapy , Osteogenesis , Silicates/chemistry , Tissue Engineering , Tissue Scaffolds , Animals , Ceramics/chemistry , Femoral Fractures/etiology , Femoral Fractures/pathology , Mechanical Phenomena , Porosity , Rabbits
12.
FASEB J ; 34(4): 5208-5222, 2020 04.
Article in English | MEDLINE | ID: mdl-32060985

ABSTRACT

Emerging evidence highlights the role of the long noncoding RNA (lncRNA) KCNQ1OT1 in fracture healing. Osteoblast proliferation, migration, and survival are pivotal during this process. In this study, we aimed to improve our understanding of the regulatory role of lncRNA KCNQ1OT1 during osteoblast proliferation, migration, and survival. We searched the gene expression omnibus databases and LncBase Experimental V.2 to identify key microRNAs (miRNAs) targets of KCNQ1OT1. MiR-701-3p was selected as a differentially expressed miRNA and RNA immunoprecipitation assays were performed to verify its interaction with KCNQ1OT1. Fibroblast growth factor receptor 3 (FGFR3) was also identified as a target of miR-701-3p. We further identified KCNQ1OT1 as a competing endogenous RNA of miR-701-3p that could influence osteoblast proliferation, migration, and apoptosis in vitro and in vivo. Taken together, our results indicate that the KCNQ1OT1/miR-701-3p/FGFR3 axis is an important regulator of osteoblast proliferation, migration, and apoptosis, and provide a new therapeutic avenue for fracture healing.


Subject(s)
Disease Models, Animal , Femoral Fractures/therapy , Fracture Healing/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Animals , Apoptosis , Cell Proliferation , Femoral Fractures/pathology , Male , Mice , Mice, Inbred C57BL , Receptor, Fibroblast Growth Factor, Type 3/genetics , Signal Transduction
13.
Cell Biol Int ; 45(1): 83-91, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32991030

ABSTRACT

microRNAs (miRNAs) have recently been recognized as playing an important role in bone-associated diseases. This study investigated whether the reduced miR-155-5p in steroid-associated osteonecrosis of the femoral head (ONFH) attenuated osteogenic differentiation and cell proliferation by targeting GSK3B. Bone marrow was collected from the proximal femurs of patients with steroid-associated ONFH (n = 10) and patients with new femoral neck fracture (n = 10) and mesenchymal stem cells (MSCs) were isolated. The expression profile, the biological function of miR-155-5p, and the interaction between miR-155-5p and GSK3B were investigated by cell viability measurement, western blot, real-time polymerase chain reaction, luciferase reporter assay, and Alizarin Red S (ARS) staining of MSCs. The MSCs that were obtained from the femoral neck fracture group and from the steroid-associated ONFH group were transfected with or without miR-155-5p. We found that, in ONFH samples, the level of mature miR-155-5p was significantly lower than that of control samples. By inhibiting GSK3B, miR-155-5p promoted the nuclear translocation of ß-catenin, increased the expression of osteogenesis-related genes, and facilitated the proliferation and differentiation of MSCs. Restoring the expression of GSK3B in MSCs partially reversed the effect of miR-155-5p. These findings suggest that reduced miR-155-5p in steroid-associated ONFH attenuates osteogenic differentiation and cell proliferation by increased levels of GSK3B and inhibition of Wnt signaling.


Subject(s)
Femur Head Necrosis/chemically induced , Femur Head Necrosis/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Osteogenesis/genetics , Steroids/adverse effects , Base Sequence , Cell Nucleus/metabolism , Cell Proliferation/genetics , Female , Femoral Fractures/genetics , Femoral Fractures/pathology , Humans , Male , MicroRNAs/genetics , Middle Aged , Protein Transport , Signal Transduction , beta Catenin/metabolism
14.
Proc Natl Acad Sci U S A ; 115(50): E11741-E11750, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30478052

ABSTRACT

A comprehensive understanding of the key microenvironmental signals regulating bone regeneration is pivotal for the effective design of bioinspired orthopedic materials. Here, we identified citrate as an osteopromotive factor and revealed its metabonegenic role in mediating citrate metabolism and its downstream effects on the osteogenic differentiation of human mesenchymal stem cells (hMSCs). Our studies show that extracellular citrate uptake through solute carrier family 13, member 5 (SLC13a5) supports osteogenic differentiation via regulation of energy-producing metabolic pathways, leading to elevated cell energy status that fuels the high metabolic demands of hMSC osteodifferentiation. We next identified citrate and phosphoserine (PSer) as a synergistic pair in polymeric design, exhibiting concerted action not only in metabonegenic potential for orthopedic regeneration but also in facile reactivity in a fluorescent system for materials tracking and imaging. We designed a citrate/phosphoserine-based photoluminescent biodegradable polymer (BPLP-PSer), which was fabricated into BPLP-PSer/hydroxyapatite composite microparticulate scaffolds that demonstrated significant improvements in bone regeneration and tissue response in rat femoral-condyle and cranial-defect models. We believe that the present study may inspire the development of new generations of biomimetic biomaterials that better recapitulate the metabolic microenvironments of stem cells to meet the dynamic needs of cellular growth, differentiation, and maturation for use in tissue engineering.


Subject(s)
Citric Acid/metabolism , Mesenchymal Stem Cells/metabolism , Adenosine Triphosphate/metabolism , Animals , Biocompatible Materials/chemistry , Biopolymers/chemistry , Bone Regeneration/physiology , Cell Adhesion , Cell Differentiation/physiology , Cell Proliferation , Disease Models, Animal , Femoral Fractures/pathology , Femoral Fractures/therapy , Humans , Male , Mesenchymal Stem Cells/cytology , Metabolic Networks and Pathways , Models, Biological , Osteogenesis/physiology , Phenotype , Phosphoserine/metabolism , Rats , Rats, Sprague-Dawley , Skull Fractures/pathology , Skull Fractures/therapy , Stem Cell Niche/physiology , Symporters/metabolism , Tissue Engineering , Tissue Scaffolds/chemistry
15.
Wilderness Environ Med ; 32(1): 59-62, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33339718

ABSTRACT

Although stress injuries are a common occurrence in sports medicine clinics, a distal femur stress fracture is less so. Early detection can result in a favorable prognosis and may prevent the need for surgical intervention. A misdiagnosis resulting in delay of care can result in significant complications. This case report documents a rare distal femur stress fracture in a long-distance hiker. A 35-y-old male presented to an orthopedic clinic in Pennsylvania with left knee pain after completing 1423 km (884 mi) of the Appalachian trail over a 4-mo period. He was attempting a thru-hike, a specialized type of backpacking focused on completing a trail from end-to-end. Thru-hiking of this trail involves backpacking between Georgia and Maine, covering about 3540 km (2200 mi) with approximately 141,580 m (464,500 ft) of gain/loss in elevation. His pain began 2 mo into his hike when he noted medial sided left knee discomfort. Over the following 2 mo he sought treatment at 2 different locations along the trail with etiology undetermined. Upon evaluation in Pennsylvania, history and physical exam were suggestive of a stress fracture. Radiologic studies confirmed a closed nondisplaced nonangulated grade 4 transverse fracture of the shaft of the distal left femur. The patient was instructed to terminate his hike immediately and he was placed on nonweight bearing status. This case illustrates the importance of considering a distal femur stress fracture for the differential diagnosis of persistent knee pain in a long-distance hiker.


Subject(s)
Femoral Fractures/diagnosis , Femur/pathology , Fractures, Stress/diagnosis , Adult , Femoral Fractures/pathology , Humans , Male , Sports
16.
J Zoo Wildl Med ; 52(2): 564-572, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34130399

ABSTRACT

A retrospective study was performed to describe patient and fracture characteristics, treatments, complications, and outcomes associated with femoral fractures in free-ranging birds of prey. Cases were identified from the electronic database and included raptors admitted with femoral fractures at the Bird of Prey Clinic of the Université de Montréal, from 1986 to 2018. In total, 119 individuals from 24 species, with 123 femoral fractures, were included in the study. Twenty-seven birds (23%) were declared dead upon arrival. Ninety-two birds (77%) were admitted alive; 80 with acute fractures and 12 with a fracture that had already healed. A conservative treatment (cage rest) was used to manage eight fractures. Surgical repair was attempted on 34 femoral fractures in 33 birds. An intramedullary pin alone was used in 28 cases (82%), and an external skeletal fixator-intramedullary pin tie-in fixator was used in 6 cases (18%). For surgically treated fractures, 19 healed (in 18 birds), 4 did not heal, and 11 birds died or were euthanized before healing could be achieved. Of the 23/34 fractures where the healing process could be assessed, healing rate for comminuted fractures (11/23) was 91% (10/11). Surgical complications occurred in 10 cases (including 7 cases of migration and loosening of the pin, and 1 case of osteomyelitis), leading to euthanasia in 4 cases. Out of the 21 birds with acute femoral fracture upon presentation that were released, 14 had received a surgical treatment and 7 had received a conservative treatment. Nine of the 21 released birds (43%) had at least one other fractured bone. The majority of deaths and euthanasia occurred within 2 wk after admission (51/59; 86%). Thirteen birds were euthanized primarily because of their femoral fracture. Thirteen birds died in treatment and 33 were deemed nonreleasable and euthanized because of a comorbidity not related to the femoral fracture.


Subject(s)
Bird Diseases/etiology , Femoral Fractures/veterinary , Hindlimb/pathology , Raptors/injuries , Animals , Bird Diseases/pathology , Bird Diseases/therapy , Femoral Fractures/pathology , Femoral Fractures/therapy , Retrospective Studies , Treatment Outcome
17.
Biochem Biophys Res Commun ; 530(4): 632-637, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32762942

ABSTRACT

Extracorporeal shock wave therapy (ESWT) has been demonstrated to accelerate bone healing; however, the mechanism underlying ESWT-induced bone regeneration has not been fully elucidated. This study aimed to examine the effects of ESWT and the process of fracture healing. A rat model of femur delayed-union was established by cauterizing the periosteum. ESWT treatment at the fracture site was performed 2 weeks after the operation and the site was radiographically and histologically evaluated at weeks 4, 6, and 8. The bone union rate and radiographic score of the ESWT group were significantly higher than those of the control group at 8 weeks. Histological evaluation revealed enhanced endochondral ossification at the fracture site. The effects of ESWT on ATDC5 cells were examined in vitro. ESWT promoted chondrogenic differentiation without inhibiting the proliferation of ATDC5 cells. ESWT may induce significant bone healing by promoting endochondral ossification at the fracture site.


Subject(s)
Extracorporeal Shockwave Therapy , Femoral Fractures/therapy , Femur/injuries , Fracture Healing , Osteogenesis , Animals , Bone Regeneration , Cell Line , Cell Proliferation , Disease Models, Animal , Extracorporeal Shockwave Therapy/methods , Femoral Fractures/pathology , Femoral Fractures/physiopathology , Femur/pathology , Femur/physiopathology , Male , Mice , Rats , Rats, Sprague-Dawley
18.
Biochem Biophys Res Commun ; 525(4): 850-856, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32169280

ABSTRACT

AIM: To investigate the effect of endogenous PTH deficiency on osteoclasts during fracture healing and its mechanism. METHODS: A femoral fracture model was used to determine the role of endogenous PTH in fracture healing. Immunohistochemistry, qPCR, and Western blot were used to determine the potential functions and mechanisms of endogenous PTH. RESULT: In this study, we found that expression of RANKL and CK was lower in PTH knockout (KO) mice than in wild type (WT) mice. In vitro culture of osteoclasts showed that under the same stimulation, there was no statistical difference in the number of osteoclasts and the area of bone resorption areas in PTH WT mice and PTH KO mice. We found that a high concentration of RANKL could promote the number and activity of osteoclasts. Upon induction of osteoblasts in vitro, those from the PTH WT group expressed higher RANKL protein and mRNA than those from the PTH KO group. Lastly, we confirmed that the PI3K/AKT/STAT5 pathway promotes RANKL increase from osteoblasts. CONCLUSION: During fracture healing, endogenous PTH deficiency can affect osteoclast activity by reducing RANKL expression in osteoblasts.


Subject(s)
Fracture Healing/physiology , Osteoblasts/metabolism , Osteoclasts/metabolism , Parathyroid Hormone/metabolism , RANK Ligand/metabolism , Animals , Cell Communication , Cells, Cultured , Femoral Fractures/metabolism , Femoral Fractures/pathology , Mice, Knockout , Parathyroid Hormone/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism
19.
Am J Pathol ; 189(1): 147-161, 2019 01.
Article in English | MEDLINE | ID: mdl-30339839

ABSTRACT

The terminal complement complex (TCC) is formed on activation of the complement system, a crucial arm of innate immunity. TCC formation on cell membranes results in a transmembrane pore leading to cell lysis. In addition, sublytic TCC concentrations can modulate various cellular functions. TCC-induced effects may play a role in the pathomechanisms of inflammatory disorders of the bone, including rheumatoid arthritis and osteoarthritis. In this study, we investigated the effect of the TCC on bone turnover and repair. Mice deficient for complement component 6 (C6), an essential component for TCC assembly, and mice with a knockout of CD59, which is a negative regulator of TCC formation, were used in this study. The bone phenotype was analyzed in vivo, and bone cell behavior was analyzed ex vivo. In addition, the mice were subjected to a femur osteotomy. Under homeostatic conditions, C6-deficient mice displayed a reduced bone mass, mainly because of increased osteoclast activity. After femur fracture, the inflammatory response was altered and bone formation was disturbed, which negatively affected the healing outcome. By contrast, CD59-knockout mice only displayed minor skeletal alterations and uneventful bone healing, although the early inflammatory reaction to femur fracture was marginally enhanced. These results demonstrate that TCC-mediated effects regulate bone turnover and promote an adequate response to fracture, contributing to an uneventful healing outcome.


Subject(s)
Bone Regeneration , Complement Membrane Attack Complex , Femoral Fractures , Fracture Healing , Osteoclasts , Animals , Bone Regeneration/genetics , Bone Regeneration/immunology , CD59 Antigens/deficiency , Cell Culture Techniques , Complement C6/deficiency , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/immunology , Complement Membrane Attack Complex/metabolism , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/pathology , Femoral Fractures/genetics , Femoral Fractures/immunology , Femoral Fractures/metabolism , Femoral Fractures/pathology , Fracture Healing/genetics , Fracture Healing/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Osteoclasts/immunology , Osteoclasts/metabolism , Osteoclasts/pathology , Sheep
20.
Calcif Tissue Int ; 106(2): 172-179, 2020 02.
Article in English | MEDLINE | ID: mdl-31578632

ABSTRACT

Clinical concerns have been raised over prior exposure to bisphosphonates impairing fracture healing. To model this, groups of male Wistar rats were assigned to saline control or treatment groups receiving 0.15 mg/kg (low dose), 0.5 mg/kg (medium dose), and 5 mg/kg (high dose) Pamidronate (PAM) twice weekly for 4 weeks. At this point, closed fractures were made using an Einhorn apparatus, and bisphosphonate dosing was continued until the experimental endpoint. Specimens were analyzed at 2 and 6 weeks (N = 8 per group per time point). Twice weekly PAM dosing was found to have no effect on early soft callus remodeling at 2 weeks post fracture. At this time point, the highest dose PAM group gave significant increases in bone volume (+ 10%, p < 0.05), bone mineral content (+ 30%, p < 0.01), and bone mineral density (+ 10%, p < 0.01). This PAM dosing regimen showed more substantive effects on hard callus at 6 weeks post fracture, with PAM treatment groups showing + 46-79% increased bone volume. Dynamic bone labeling showed reduced calcein signal in the PAM-treated calluses (38-63%, p < 0.01) and reduced MAR (32-49%, p < 0.01), suggesting a compensatory reduction in bone anabolism. These data support the concept that bisphosphonates lead to profound decreases in bone turnover in fracture repair, however, this does not affect soft callus remodeling.


Subject(s)
Bony Callus/drug effects , Femoral Fractures/pathology , Fractures, Closed/pathology , Osteogenesis/drug effects , Pamidronate/pharmacology , Animals , Bone Density/drug effects , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , Bony Callus/pathology , Disease Models, Animal , Down-Regulation/drug effects , Femoral Fractures/drug therapy , Fracture Healing/drug effects , Fractures, Closed/drug therapy , Male , Organ Size/drug effects , Osteogenesis Imperfecta/pathology , Pamidronate/administration & dosage , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL