Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
J Pediatr Orthop ; 40(6): e473-e478, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32501918

ABSTRACT

BACKGROUND: Arthropathies and bone deformities are well known to occur in patients with thalassemia major and have been attributed to the disease or to its therapy. Before the advent of chelation therapy, these children developed widened diploic space and "hair-on-end" pattern in skull, "cobweb" pattern in the pelvis, and the lack of the normal concave outline in the long bones because of extensive marrow proliferation. After the introduction of iron-chelation therapy, these patients were noted to develop metaphyseal abnormalities and vertebral changes resembling spondylo-metaphyseal dysplasia. Only one study has shown some association of deferiprone (chelating agent) use with distal ulnar changes in these children. Our study was done to describe the skeletal changes and deformities in wrist joints of children with transfusion-dependent thalassemia and correlate them with age, mean pretransfusion hemoglobin level, mean serum ferritin level, and type and duration of chelation therapy in these children. METHODS: A total of 60 children with transfusion-dependent thalassemia from the thalassemia daycare center were examined. These children were divided into 3 groups on the basis of their age (group A: 2 to 6 y, group B: 6 to 10 y, and group C: 10 to 14 y). Detailed history, including treatment history, number of blood transfusions received over the last 1 year, clinical examination, and radiologic assessment of both forearm with wrists were done. RESULTS: The clinical and radiologic differences in radial and ulnar lengths increased significantly with the increasing age of these patients, the ulna being short. There was some correlation between increasing negative ulnar variance and distal radial articular angle with deferiprone consumption. CONCLUSION: Chelation therapy, particularly with deferiprone, may cause distal ulnar growth arrest causing ulnar shortening and progressive radial bowing in these children. LEVEL OF EVIDENCE: Level IV-case series.


Subject(s)
Chelation Therapy/adverse effects , Deferiprone/adverse effects , Iron Chelating Agents/adverse effects , Wrist Joint/drug effects , beta-Thalassemia/drug therapy , Adolescent , Blood Transfusion , Child , Child, Preschool , Female , Forearm/diagnostic imaging , Humans , Joint Diseases/etiology , Male , Radiography , Radius/diagnostic imaging , Radius/drug effects , Ulna/diagnostic imaging , Ulna/drug effects , Wrist/diagnostic imaging , Wrist Joint/diagnostic imaging
2.
Biomed Tech (Berl) ; 65(4): 491-505, 2020 Aug 27.
Article in English | MEDLINE | ID: mdl-32335536

ABSTRACT

The aim of this study was to develop novel hydroxyapatite (HAP)-based bioactive bone replacement materials for segmental osteotomy reconstruction. Customized three-dimensional (3D) bone construct was manufactured from nanohydroxyapatite (nHAP) with poly(lactide-co-glycolide) (PLGA) coating using 3D models derived from the computed tomography (CT) scanning of the rabbit's ulna and gradient 3D printing of the bone substitute mimicking the anatomical shape of the natural bone defect. Engineered construct revealed adequate micro-architectural design for successful bone regeneration having a total porosity of 64% and an average pore size of 256 µm. Radiography and micro-CT analysis depicted new bone apposition through the whole length of the reconstructed ulna with a small area of non-resorbed construct in the central area of defect. Histological analysis revealed new bone formation with both endochondral and endesmal type of ossification. Immunohistochemistry analysis depicted the presence of bone formation indicators - bone morphogenetic protein (BMP), osteocalcin (OCN) and osteopontin (OPN) within newly formed bone. Manufactured personalized construct acts as a "smart" responsive biomaterial capable of modulating the functionality and potential for the personalized bone reconstruction on a clinically relevant length scale.


Subject(s)
Bone Regeneration/physiology , Bone Substitutes/chemistry , Osteogenesis/physiology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Ulna/chemistry , Animals , Biocompatible Materials/chemistry , Biomimetics , Durapatite/chemistry , Printing, Three-Dimensional , Rabbits , Tissue Engineering/methods , Ulna/drug effects
3.
Arthritis Res Ther ; 21(1): 174, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31307521

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is known to cause secondary osteoporosis and fragility fractures. This study aimed to identify biomarkers predictive of bone mineral density (BMD) change at three anatomical sites in patients with RA. METHODS: We conducted a prospective longitudinal study in patients with RA. In 2012, we recruited 379 patients from an RA cohort, 329 of whom underwent evaluation of blood and urine biomarkers together with measurement of BMD in the lumbar spine, proximal femur, and distal forearm. The BMD in these three regions was reassessed in 2014. We performed multivariate linear regression analysis to identify those factors associated with BMD change. RESULTS: The averages of age, body mass index, and disease activity score in 28 joints (DAS28) at baseline were 63.2 (minimum to maximum, 32-85), 21.3 (12.3-30.0), and 3.2 (0.1-5.9), respectively. Univariate analysis showed that the annual BMD change was significantly associated with the use of steroid, bisphosphonate (BP) or vitamin D (VitD), and serum homocysteine in the lumber spine; DAS28, the use of BP or VitD, CRP, and anti-cyclic citrullinated peptide antibody (ACPA) in the proximal femur; and the dosage of MTX, the use of BP or VitD, and serum tartrate-resistant acid phosphatase 5b (TRACP-5b) in the distal forearm, respectively. CONCLUSIONS: Predictive biomarkers for BMD change in RA patients differ at each anatomical site. Practitioners should treat each anatomical site with different markers and prescribe osteoporosis drugs to prevent fractures for RA patients.


Subject(s)
Arthritis, Rheumatoid/metabolism , Biomarkers/analysis , Bone and Bones/metabolism , Osteoporosis/metabolism , Absorptiometry, Photon/methods , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/drug therapy , Biomarkers/blood , Biomarkers/urine , Bone Density/drug effects , Bone Density Conservation Agents/therapeutic use , Bone and Bones/drug effects , Diphosphonates/therapeutic use , Female , Femur/drug effects , Femur/metabolism , Humans , Longitudinal Studies , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/metabolism , Male , Middle Aged , Osteoporosis/diagnosis , Osteoporosis/drug therapy , Prospective Studies , Radius/drug effects , Radius/metabolism , Ulna/drug effects , Ulna/metabolism , Vitamin D/therapeutic use
4.
Indian Pediatr ; 56(1): 41-44, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30806360

ABSTRACT

OBJECTIVE: To describe a novel wrist deformity in b-thalassemia major patients, and their radiographic and magnetic resonance imaging findings. METHODS: 30 patients with b-thalassemia major who were noticed to have ulnar deviation at wrist joint were evaluated for previous history of medications, serum ferritin levels, presence of pain and swelling at the wrist joint, and the duration of iron chelation therapy. Radiographs of wrist and limited magnetic resonance imaging (MRI) sequences were obtained in 30 and 15 patients, respectively. RESULTS: Radiographs revealed varying severity of distal ulnar shortening, distal radial slanting and presence of soft tissue distal to the ulna. MRI showed similar deformities along with abnormal marrow signal at distal ulnar ends; in 8 patients, a soft tissue distal to the distal end of ulna was noted. CONCLUSIONS: Varying severity of radiological abnormalities, predominantly affecting the distal ulna, are present in children and adolescents with b-thalassemia receiving oral chelation therapy.


Subject(s)
Chelation Therapy/adverse effects , Iron Chelating Agents/adverse effects , Joint Diseases , Wrist , beta-Thalassemia/drug therapy , Adolescent , Child , Female , Humans , Iron , Iron Chelating Agents/therapeutic use , Joint Diseases/chemically induced , Joint Diseases/diagnostic imaging , Joint Diseases/pathology , Male , Ulna/diagnostic imaging , Ulna/drug effects , Ulna/pathology , Wrist/diagnostic imaging , Wrist/pathology
5.
Hand Surg Rehabil ; 38(1): 71-73, 2019 02.
Article in English | MEDLINE | ID: mdl-30401614

ABSTRACT

We report the case of a 28-year-old man with a septic forearm non-union treated with minocycline for 3 months. At the time of reconstructive surgery, the radius and ulna were entirely black. Surgical debridement until bleeding of both bone extremities resulted in a 5-cm defect that was filled with a cement spacer. Histology confirmed poorly vascularized bone with focal areas of acute inflammatory infiltrate at the non-union sites (highly suggestive of infection) and normal structure of the remaining diaphyseal bones, although black in color. Reconstruction with free vascularized fibula transfer was successful leading to complete bone healing. An incidental finding of minocycline-induced black bone discoloration should not change the surgeon's decision because there is no evidence of adverse effects on bone healing in the literature. Surgery can be performed safely at sites of minocycline-induced black bone pigmentation.


Subject(s)
Anti-Bacterial Agents/adverse effects , Minocycline/adverse effects , Pigmentation Disorders/chemically induced , Radius/drug effects , Ulna/drug effects , Adult , Anti-Bacterial Agents/administration & dosage , Fracture Fixation, Intramedullary/adverse effects , Fractures, Ununited/surgery , Humans , Male , Minocycline/administration & dosage , Postoperative Complications , Radius/pathology , Radius Fractures/surgery , Reoperation , Surgical Wound Infection/drug therapy , Ulna/pathology , Ulna Fractures/surgery
6.
J Biomed Mater Res A ; 106(7): 1842-1850, 2018 07.
Article in English | MEDLINE | ID: mdl-29573560

ABSTRACT

We previously reported that biphasic calcium phosphate (BCP) microparticles embedded in a blood clot induces ectopic bone formation in mice and repairs a critical femoral defect in rat. The present pilot study aimed to evaluate in dog and in two models of large defects the efficacy of this composite named "blood for reconstruction of bone" (BRB). We show here that BRB is a cohesive biomaterial easy to prepare from dog autologous blood and to mold to fill large bone defects. First in a model of cylindrical femoral condyle defect, the BRB was compared with BCP particles alone. After 8 weeks, this revealed that the amount of mature bone was slightly and significantly higher with BRB than with BCP particles. Second, in a model consisting in a 2 cm-long critical interruptive defect of the ulna, the BRB was compared with autologous bone. After 6 months, we observed that implantation of BRB can induce the complete reconstruction of the defect and that newly formed bone exhibits high regenerative potential. Comparison with the results obtained with autologous bone grafting strongly suggests that the BRB might be an efficient biomaterial to repair large bone defects, as an alternative or in addition to autologous bone. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1842-1850, 2018.


Subject(s)
Blood/metabolism , Bone and Bones/drug effects , Bone and Bones/pathology , Calcium Phosphates/pharmacology , Microspheres , Animals , Bone Regeneration/drug effects , Dogs , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Implants, Experimental , Osteogenesis/drug effects , Pilot Projects , Ulna/diagnostic imaging , Ulna/drug effects , Ulna/pathology , X-Ray Microtomography
7.
Bone ; 105: 67-74, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28826844

ABSTRACT

Small animal models, and especially transgenic models, have become widespread in the study of bone mechanobiology and metabolic bone disease, but test methods for measuring fracture toughness on multiple replicates or at multiple locations within a single small animal bone are lacking. Therefore, the objective of this study was to develop a method to measure cortical bone fracture toughness in multiple specimens and locations along the diaphysis of small animal bones. Arc-shaped tension specimens were prepared from the mid-diaphysis of rabbit ulnae and loaded to failure to measure the radial fracture toughness in multiple replicates per bone. The test specimen dimensions, crack length, and maximum load met requirements for measuring the plane strain fracture toughness. Experimental groups included a control group, bisphosphonate treatment group, and an ex vivo deproteinization treatment following bisphosphonate treatment (5 rabbits/group and 15 specimens/group). The fracture toughness of ulnar cortical bone from rabbits treated with zoledronic acid for six months exhibited no difference compared with the control group. Partially deproteinized specimens exhibited significantly lower fracture toughness compared with both the control and bisphosphonate treatment groups. The deproteinization treatment increased tissue mineral density (TMD) and resulted in a negative linear correlation between the measured fracture toughness and TMD. Fracture toughness measurements were repeatable with a coefficient of variation of 12-16% within experimental groups. Retrospective power analysis of the control and deproteinization treatment groups indicated a minimum detectable difference of 0.1MPa·m1/2. Therefore, the overall results of this study suggest that arc-shaped tension specimens offer an advantageous new method for measuring the fracture toughness in small animal bones.


Subject(s)
Cortical Bone/physiopathology , Diphosphonates/therapeutic use , Fractures, Bone/drug therapy , Fractures, Bone/physiopathology , Proteins/isolation & purification , Animals , Biomechanical Phenomena/drug effects , Bone Density/drug effects , Cortical Bone/drug effects , Cortical Bone/pathology , Diphosphonates/pharmacology , Imaging, Three-Dimensional , Male , Rabbits , Ulna/diagnostic imaging , Ulna/drug effects , Ulna/physiopathology , X-Ray Microtomography
8.
Arthritis Rheumatol ; 69(9): 1741-1750, 2017 09.
Article in English | MEDLINE | ID: mdl-28544807

ABSTRACT

OBJECTIVE: Articular erosions correlate with disability in rheumatoid arthritis (RA). Biologic agents reduce erosion progression in RA, but erosion healing occurs infrequently. This study was undertaken to assess the effects of the anabolic agent teriparatide on joint erosion volume in RA patients treated with a tumor necrosis factor inhibitor (TNFi). METHODS: We conducted a randomized controlled trial in 24 patients with erosive RA, osteopenia, and disease activity controlled by TNFi treatment for at least 3 months. Half were randomized to receive teriparatide for 1 year and the others constituted a wait-list control group. Subjects and primary rheumatologists were not blinded with regard to treatment assignment, but all outcomes were assessed in a blinded manner. The primary outcome measure was change in erosion volume determined by computed tomography at 6 anatomic sites. Significance within each hand and anatomic site was based on a 2-tailed test, with P values less than 0.05 considered significant. RESULTS: Baseline characteristics of the treatment groups were well balanced. After 52 weeks, the median change in erosion volume in the teriparatide group was -0.4 mm3 (interquartile range [IQR] -34.5, 29.6) and did not differ significantly from that in controls (median change +9.1 mm3 [IQR -29.6, 26.4]) (P = 0.28). No significant difference in change in erosion volume was noted at the radius, ulna, or metacarpophalangeal joints. Bone mineral density improved at the femoral neck and lumbar spine in the teriparatide group. CONCLUSION: Our findings indicate that teriparatide treatment for 1 year does not significantly reduce erosion volume in the hands or wrists of patients with established RA with disease activity controlled by TNFi treatment.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Bone Density Conservation Agents/administration & dosage , Bone Density/drug effects , Bone Diseases, Metabolic/drug therapy , Teriparatide/administration & dosage , Aged , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/diagnostic imaging , Bone Diseases, Metabolic/diagnostic imaging , Bone Diseases, Metabolic/etiology , Female , Humans , Male , Metacarpophalangeal Joint/diagnostic imaging , Metacarpophalangeal Joint/drug effects , Middle Aged , Radius/diagnostic imaging , Radius/drug effects , Severity of Illness Index , Tomography, X-Ray Computed , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Ulna/diagnostic imaging , Ulna/drug effects
9.
Int J Biol Macromol ; 101: 630-637, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28363647

ABSTRACT

This study compared the regeneration potentials of the hydroxyapatite (HA) and beta-tricalcium phosphate (ß-TCP) alone or in combination with a HA:TCP ratio of 30:70 in the critical-sized radial bone defects of rats. Bilateral 60 radial bone defects created were randomly divided into six equal groups (n=10 defects/group) including autograft, untreated or defect, chitosan-gelatin-platelet gel (CGP), CGP-HA, CGP-TCP and CGP-HA/TCP. The defects were evaluated by radiography, morphology, histopathology, histomorphometry, CT scan, scanning electron microscopy and biomechanical testing after eight weeks. Compared with the untreated and CGP-HA groups, the CGP and CGP-HA/TCP groups showed significantly higher new bone formation, bone volume, and mechanical properties. The CGP-HA and CGP-TCP scaffolds showed low biodegradability, whereas the CGP scaffolds were completely degraded. Osteoconductivity and osteoinductivity of the CGP and CGP-HA/TCP scaffolds were superior to the CGP-HA ones. The untreated and CGP-HA groups repaired mostly through fibrosis, while there were evidence of higher bone formation in the autograft, CGP and CGP-HA/TCP groups. In conclusion, addition of HA or ß-TCP alone into the CGP scaffolds impaired bone regeneration, while bone regeneration with the CGP and CGP-HA/TCP scaffolds was comparable with the autograft. Therefore, the CGP-HA/TCP scaffold can be a possible option to substitute the autologous bone grafting.


Subject(s)
Blood Platelets/chemistry , Bone Regeneration/drug effects , Calcium Phosphates/chemistry , Chitosan/chemistry , Durapatite/chemistry , Gelatin/chemistry , Radius/drug effects , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biomechanical Phenomena , Male , Materials Testing , Radius/physiology , Radius/ultrastructure , Rats , Rats, Sprague-Dawley , Ulna/drug effects , Ulna/physiology , Ulna/ultrastructure
10.
Singapore Med J ; 58(9): 551-556, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27193079

ABSTRACT

INTRODUCTION: This study evaluates the effect of autologous platelet-rich plasma (PRP) combined with xenogeneic demineralised bone matrix (DBM) on bone healing of critical-size ulnar defects (2-2.5 times the ulnar diameter) in New Zealand White rabbits. METHODS: Critical-size defects were created unilaterally in the ulna of 36 rabbits, while keeping the contralateral limb intact. They were divided into three groups. In Group A, the defect was filled with autologous PRP and in Group B, with autologous PRP combined with DBM; in Group C, the defect remained empty. The rabbits were euthanised 12 weeks postoperatively. Radiological, biomechanical and histological assessments were carried out and statistical analysis of the results was performed. RESULTS: Group B had significantly higher radiological and histological scores than Groups A and C. Defects in Group B showed significant new bone formation, whereas there was minimal or no new bone formation in Groups A and C. Only specimens in Group B showed macroscopic bone union. Biomechanical evaluation of the treated and intact contralateral limbs in Group B showed significant differences. CONCLUSION: In this study, statistically significant enhancement of bone healing was found in critical-size defects treated with PRP and DBM, as shown by radiological findings, gross assessment, and biomechanical and histopathological results. Defects in the two other groups remained unbridged. Therefore, PRP was effective only when it was used in combination with a bone graft.


Subject(s)
Bone Matrix/chemistry , Bone Transplantation , Bone and Bones/drug effects , Platelet-Rich Plasma/metabolism , Ulna/drug effects , Animals , Biomechanical Phenomena , Humans , Rabbits , Transplantation, Heterologous
11.
BMC Vet Res ; 12: 201, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27619812

ABSTRACT

BACKGROUND: Large bone defects in canines usually require assistance to achieve healing. Implantation of osteoinductive factors can promote bone healing, while transplantation of osteoprogenitor cells can enhance bone regeneration. We hypothesized that implantation of an osteoinductive factor, recombinant human bone morphogenetic protein-2 (rhBMP-2), combined with osteoprogenitor cells, bone marrow-derived mesenchymal stromal cells (BMSCs), would synergistically promote bone healing. In this study, we examined the combined effects of Escherichia coli-derived rhBMP-2 and BMSCs on bone healing after implantation into canine ulnar defects. RESULTS: Critical-sized osteoperiosteal segmental defects (2.5 cm) were created in the ulnae of healthy female beagle dogs, and implanted with combinations of E. coli-derived rhBMP-2 (560 or 140 µg) and autologous BMSCs (10(7), 10(5), or 0 cells). In the present study,18 forelimbs of nine healthy purpose-bred female beagles were used. All six treatment groups contained three forelimbs, and the animals were euthanized after 12 weeks. The control groups (560 and 140 µg/0 cells) were cited from our previous study to reduce the number of experimental animals. Radiographically, the regenerated bone width was significantly increased in the 560 or 140 µg with 10(7) and 10(5) cells groups compared with the 0 cells groups. By quantitative CT, the bone mineral density was higher in the 560 µg with 10(7) and 10(5) cells groups, while non-uniformity of the bone mineral density was improved in the 560 µg with 10(7) and 10(5) cells groups and 140 µg/10(7) cells group. Mechanically, the maximum loads at failure were significantly higher in the 560 µg with 10(7) and 10(5) cells groups. Histologically, the regenerated bone was well-developed and contained osteocyte-like cells marrow cavities, and vessels. However, the osteoclasts and osteoblasts were hardly observed. The osteocyte-like cell numbers were significantly higher in the 560 µg with 10(7) and 10(5) cells and 140 µg with 10(7) and 10(5) cells groups. CONCLUSIONS: Implantation of E. coli-derived rhBMP-2 and BMSCs led to significantly enhanced bone formation, with improved bone mineral density and reduced non-uniformity of the regenerated bone. Combined implantation of rhBMP-2 and BMSCs may be useful for promotion of bone healing in critical-sized defects in canines.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Dog Diseases/therapy , Escherichia coli/metabolism , Mesenchymal Stem Cell Transplantation/veterinary , Recombinant Proteins/metabolism , Ulna/injuries , Animals , Biocompatible Materials/therapeutic use , Bone Density/drug effects , Bone Density Conservation Agents/therapeutic use , Bone Marrow Cells/physiology , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Regeneration , Calcium Phosphates/therapeutic use , Dogs , Escherichia coli/genetics , Female , Humans , Implants, Experimental , Mesenchymal Stem Cells/physiology , Recombinant Proteins/genetics , Ulna/drug effects
12.
Int J Surg Pathol ; 24(4): 366-72, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26842345

ABSTRACT

Denosumab has shown promising results in the management of giant cell tumor of bone, a primary bone tumor with locally aggressive behaviour. We report a case of premature denosumab interruption due to radiological and clinical tumor expansion of a giant cell tumor of the distal ulna. Although denosumab is known to induce tumor regression, with progressive ossification and loss of the characteristic morphology of giant cell tumor of bone, the ulnar tumor specimen showed a moderately to highly cellular proliferation of short spindle-shaped cells, and no osteoclast-like giant cells. There were no abnormal mitotic figures. We considered the surgical specimen as a giant cell tumor of bone with partial regression after prematurely interrupted denosumab treatment. This case illustrates the diagnostic issues of an initially unfavourable evolution raising concern for malignancy, and the difficulties in histological assessment of a partially treated giant cell tumor of bone, that may mimic osteosarcoma.


Subject(s)
Antineoplastic Agents/administration & dosage , Bone Neoplasms/pathology , Denosumab/administration & dosage , Giant Cell Tumor of Bone/pathology , Ulna/drug effects , Adult , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Diagnosis, Differential , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/genetics , Histones/metabolism , Humans , Male , Osteosarcoma/diagnosis , Osteosarcoma/pathology , Polymerase Chain Reaction , Ulna/pathology
13.
Tissue Eng Part A ; 22(7-8): 610-20, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26914219

ABSTRACT

Despite recent progress, segmental bone defect repair is still a significant challenge in orthopedic surgery. While bone tissue engineering approaches using biodegradable matrices along with bone/blood vessel forming cells offered improved possibilities, current regenerative strategies lack the ability to achieve vascularized bone regeneration in critical-sized/segmental bone defects. In this study, we introduced and evaluated a two-pronged approach for vascularized bone regeneration in vivo. The goal was to demonstrate vascularized bone formation using oxygen tension-controlled (OTC) matrices seeded with bone and blood vessel forming cells. OTC matrices were coimplanted with rabbit mesenchymal stem cells (MSCs) and peripheral blood-derived endothelial progenitor cells (PB-EPCs) to demonstrate the osteogenic and vasculogenic differentiation of these cells, postseeding on a matrix, especially deep inside the matrix pore structure. Matrices coimplanted with varied rabbit MSC and PB-EPC ratios (1:4, 1:1, and 4:1) were assessed in a nude mouse subcutaneous implantation model to determine a coimplantation ratio with superior osteogenic as well as vasculogenic properties. The implants were analyzed, at week 8, for endothelial (CD31 and Von Willebrand factor [vWF]) and osteogenic marker (RunX2 and Col I) staining qualitatively and collagen deposition and number of vessel formation quantitatively. Results from these experiments established MSC-to-PB-EPC ratio 1:1 as the best coimplantation ratio. OTC matrix with 1:1 coimplantation ratio was assessed for segmental bone defect repair in a rabbit critical-sized bone defect model. The group under investigation was OTC matrix, and the matrix was seeded with MSCs, EPCs, or MSCs:EPCs in a 1:1 ratio. Explants at week 12 were evaluated for bone defect repair via micro-CT and histology. Results from rabbit in vivo experiments show enhanced mineralization and vascularization for the 1:1 coimplantation group. Overall, the study establishes a two-pronged approach involving OTC matrix and effective progenitors for large-area and vascularized bone regeneration.


Subject(s)
Bone Regeneration/drug effects , Endothelial Cells/cytology , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic/drug effects , Osteogenesis/drug effects , Oxygen/pharmacology , Tissue Scaffolds/chemistry , Animals , Biomarkers/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Fluorescent Antibody Technique , Implants, Experimental , Mesenchymal Stem Cells/drug effects , Mice, SCID , Rabbits , Ulna/diagnostic imaging , Ulna/drug effects , Ulna/pathology , X-Ray Microtomography
14.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 40(1): 25-30, 2015 Jan.
Article in Chinese | MEDLINE | ID: mdl-25652372

ABSTRACT

OBJECTIVE: To explore the effect of methylprednisolone on bone mass, microarchitecture and microdamage in cortical bone of ulna in rats. METHODS: Twenty female Sprague-Dawley rats (3.5 months old) were randomly assigned to two groups: a treatment group and a control group (n=10 per group). The treatment group was subcutaneously injected with methylprednisolone 3.5 mg/(kg.d) while the control group was subcutaneously injected with same volume of vehicle (saline). Rats were sacrificed at 9 weeks after the treatments. Before the sacrifice, the body weight and total bone mineral density (BMD) were measured. The right forelimb was separated through humeral shoulder and then single axial fatigue loading was performed on the right ulna. After fatigue load, the middle ulna section was bulkstained in basic fuchsin. Bone histomorphometry and microdamage analysis were performed on the middle ulna section. RESULTS: Compared with the control group, the body weight, total bone BMD and ulnas BMD in the treatment group were decreased by 15%, 6.4% and 4.3% respectively (all P<0.05); the ulna inner perimeter and marrow area in the treatment group were increased by 23.3% and 32%, respectively (both P<0.05), while the outer perimeter were decreased by 3.1% (P>0.05). There was no significant difference in the cortical and total area between the 2 groups (both P>0.05). The number of microcrack, microcrack density and microcrack surface density in the treatment group were increased by 43%, 48% and 50%, respectively, compared with those in the control group (all P<0.05), but there was no significant difference in the mean length of microcrack between the 2 groups (P>0.05). CONCLUSION: Methylprednisolone can significantly induce the bone loss and the deterioration of microarchitecture and microdamage in ulna of rats.


Subject(s)
Bone Density/drug effects , Methylprednisolone/pharmacology , Ulna/drug effects , Ulna/pathology , Animals , Female , Rats , Rats, Sprague-Dawley
15.
Acta Biomater ; 18: 262-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25712384

ABSTRACT

Each year, millions of Americans suffer bone fractures, often requiring internal fixation. Current devices, like plates and screws, are made with permanent metals or resorbable polymers. Permanent metals provide strength and biocompatibility, but cause long-term complications and may require removal. Resorbable polymers reduce long-term complications, but are unsuitable for many load-bearing applications. To mitigate complications, degradable magnesium (Mg) alloys are being developed for craniofacial and orthopedic applications. Their combination of strength and degradation make them ideal for bone fixation. Previously, we conducted a pilot study comparing Mg and titanium devices with a rabbit ulna fracture model. We observed Mg device degradation, with uninhibited healing. Interestingly, we observed bone formation around degrading Mg, but not titanium, devices. These results highlighted the potential for these fixation devices. To better assess their efficacy, we conducted a more thorough study assessing 99.9% Mg devices in a similar rabbit ulna fracture model. Device degradation, fracture healing, and bone formation were evaluated using microcomputed tomography, histology and biomechanical tests. We observed device degradation throughout, and calculated a corrosion rate of 0.40±0.04mm/year after 8 weeks. In addition, we observed fracture healing by 8 weeks, and maturation after 16 weeks. In accordance with our pilot study, we observed bone formation surrounding Mg devices, with complete overgrowth by 16 weeks. Bend tests revealed no difference in flexural load of healed ulnae with Mg devices compared to intact ulnae. These data suggest that Mg devices provide stabilization to facilitate healing, while degrading and stimulating new bone formation.


Subject(s)
Bone Plates , Bone Screws , Fracture Healing/drug effects , Magnesium/pharmacology , Ulna Fractures/pathology , Animals , Bone Development/drug effects , Materials Testing , Rabbits , Ulna/diagnostic imaging , Ulna/drug effects , Ulna/pathology , Ulna Fractures/diagnostic imaging , X-Ray Microtomography
16.
J Tissue Eng Regen Med ; 8(8): 604-11, 2014 Aug.
Article in English | MEDLINE | ID: mdl-22782937

ABSTRACT

The objective of this study was to investigate the feasibility of biodegradable gelatin-ß-tricalcium phosphate (ß-TCP) composites as a cell scaffold and controlled-release carrier of basic fibroblast growth factor (bFGF) suitable for inducing bone regeneration at a segmental bone defect. The composite of gelatin sponge and ß-TCP granules had an interconnected pore structure with an average size of 340 µm. The composite provided the controlled release of bFGF over 2 weeks. Segmental, critical-sized, bone defects of 20 mm length were created in the ulnas of New Zealand white rabbits and the gelatin-ß-TCP composites, with or without incorporated bFGF, were implanted into the defects. Bone regeneration and ß-TCP resorption profiles were evaluated by microcomputed tomography scanner analysis and haematoxylin and eosin staining. The composites incorporating bFGF promoted significantly higher bone regeneration at the defect site as compared to the bFGF-free composites. The controlled release of biologically active bFGF from the composites may possibly be achieved through the biodegradation of the composites, resulting in the promotion of bone regeneration. We conclude that the biodegradable gelatin-ß-TCP composite is a promising scaffold for bone regeneration that enables the controlled release of bFGF.


Subject(s)
Bone Regeneration/drug effects , Calcium Phosphates/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gelatin/pharmacology , Animals , Delayed-Action Preparations , Humans , Male , Microscopy, Electron, Scanning , Rabbits , Sus scrofa , Time Factors , Ulna/diagnostic imaging , Ulna/drug effects , Ulna/pathology , X-Ray Microtomography
17.
Artif Cells Nanomed Biotechnol ; 42(5): 309-15, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23899020

ABSTRACT

OBJECTIVE: To study the feasibility of therapy using nano-bone gelatin to comminuted fracture by animal experiment. METHODS: The animal models of transverse fracture were made on bilateral ulnas of 45 New Zealand white rabbits, which were divided into experimental group (repair with nano-bone gelatin), control group (repair with traditional medical glue), and blank group (unrepaired) randomly. The reconstruction effect in each group was evaluated using X-ray examination, MicroCT scanning, histopathology observation, and biomechanical test postoperation regularly. RESULT: On 12th week, the fractures in experimental and blank group get the marrow cavity completely unobstructed and good bone union; however, in control group, the fracture line slightly blurred with the marrow cavity not fully unobstructed; 6 weeks later, observation of bony calluses through MircoCT: experimental group, 68.5 ± 2.71%; blank group, 69.19 ± 2.3%; and control group, 49.35 ± 3.56%, there were no significant difference between the two groups (P > 0.05). The control group obviously showed worse bone union than the former two (P < 0.05). The histopathological examination shows that the bony calluses of experimental group are similar to those of the blank group; however, gelatin degraded slowly in control group with delayed union; on the 12th week, biomechanical test shows that the blank and experiment groups had basically same average bending strength values which had no significant difference (P > 0.05) and obviously were higher than those of the control group (P < 0.01). CONCLUSION: The nano-bone gelatin won't lead to delayed union of fractures and may be beneficial to it, and so may be an ideal gelatin for fixing small fractures.


Subject(s)
Adhesives/chemistry , Adhesives/pharmacology , Gelatin/chemistry , Gelatin/pharmacology , Nanostructures , Ulna/drug effects , Animals , Biomechanical Phenomena/drug effects , Fractures, Bone/diagnostic imaging , Fractures, Bone/pathology , Male , Organ Size/drug effects , Rabbits , Ulna/cytology , Ulna/diagnostic imaging , Ulna/pathology , X-Ray Microtomography
18.
J Appl Physiol (1985) ; 116(4): 416-24, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24356518

ABSTRACT

Despite the strong connection between angiogenesis and osteogenesis in skeletal repair conditions such as fracture and distraction osteogenesis, little is known about the vascular requirements for bone formation after repetitive mechanical loading. Here, established protocols of damaging (stress fracture) and nondamaging (physiological) forelimb loading in the adult rat were used to stimulate either woven or lamellar bone formation, respectively. Positron emission tomography was used to evaluate blood flow and fluoride kinetics at the site of bone formation. In the group that received damaging mechanical loading leading to woven bone formation (WBF), (15)O water (blood) flow rate was significantly increased on day 0 and remained elevated 14 days after loading, whereas (18)F fluoride uptake peaked 7 days after loading. In the group that received nondamaging mechanical loading leading to lamellar bone formation (LBF), (15)O water and (18)F fluoride flow rates in loaded limbs were not significantly different from nonloaded limbs at any time point. The early increase in blood flow rate after WBF loading was associated with local vasodilation. In addition, Nos2 expression in mast cells was increased in WBF-, but not LBF-, loaded limbs. The nitric oxide (NO) synthase inhibitor N(ω)-nitro-l-arginine methyl ester was used to suppress NO generation, resulting in significant decreases in early blood flow rate and bone formation after WBF loading. These results demonstrate that NO-mediated vasodilation is a key feature of the normal response to stress fracture and precedes woven bone formation. Therefore, patients with impaired vascular function may heal stress fractures more slowly than expected.


Subject(s)
Fracture Healing , Fractures, Stress/metabolism , Nitric Oxide/metabolism , Osteogenesis , Ulna Fractures/metabolism , Ulna/blood supply , Ulna/metabolism , Vasodilation , Animals , Blood Flow Velocity , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fracture Healing/drug effects , Fractures, Stress/diagnostic imaging , Fractures, Stress/physiopathology , Male , Mast Cells/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Osteogenesis/drug effects , Positron-Emission Tomography , Rats , Rats, Inbred F344 , Regional Blood Flow , Time Factors , Ulna/diagnostic imaging , Ulna/drug effects , Ulna/physiopathology , Ulna Fractures/diagnostic imaging , Ulna Fractures/pathology , Ulna Fractures/physiopathology , Vasodilation/drug effects , Weight-Bearing
19.
J Biomed Mater Res A ; 101(10): 2851-61, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23504981

ABSTRACT

Bioceramic processing using rapid prototyping technique (RPT) results in a fragile device that requires thermal treatment to improve the mechanical properties. This investigation evaluates the effect of thermal treatment on the mechanical, porosity, and bioactivity properties as well as the cytotoxicity of a porous silica-calcium phosphate nanocomposite (SCPC) implant prepared by RPT. Porous SCPC implant was subject to 3-h treatment at 800°C, 850°C, or 900°C. The compressive strength (s) and modulus of elasticity (E) were doubled when the sintering temperature is raised from 850 to 900°C measuring (s = 15.326 ± 2.95 MPa and E = 1095 ± 164 MPa) after the later treatment. The significant increase in mechanical properties takes place with minimal changes in the surface area and the percentage of pores in the range 1-356 µm. The SCPC implant prepared at 900°C was loaded with rh-BMP-2 and grafted into a segmental defect in the rabbit ulna. Histology analyses showed highly vascularized bone formation inside the defect. Histopathological analyses of the liver, spleen, kidney, heart, and the lung of rabbits grafted with and without SCPC demonstrated healthy tissues with no signs of toxicity or morphology alterations. Results of the study suggest that it is possible to engineering the mechanical properties of the SCPC implant without compromising its bioactivity. The enhanced bone formation inside the porous SCPC facilitated cell-mediated graft resorption and prohibited any accumulation of the material in the body organs.


Subject(s)
Biocompatible Materials/toxicity , Calcium Phosphates/toxicity , Implants, Experimental , Materials Testing , Silicates/toxicity , Tissue Engineering/methods , Animals , Biomechanical Phenomena/drug effects , Body Fluids , Bone Morphogenetic Protein 2/pharmacology , Cell Death/drug effects , Humans , Microscopy, Electron, Scanning , Myocardium/pathology , Porosity , Rabbits , Recombinant Proteins/pharmacology , Surface Properties , Temperature , Transforming Growth Factor beta/pharmacology , Ulna/drug effects , Ulna/pathology , Wound Healing/drug effects
20.
J Bone Joint Surg Am ; 95(6): 488-96, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23407637

ABSTRACT

BACKGROUND: Aspirin is frequently prescribed following orthopaedic surgery. Although there is substantial evidence that some nonsteroidal anti-inflammatory drugs (NSAIDs) are associated with delayed bone healing, there have been few studies of the effects of aspirin on bone healing and, to our knowledge, none on the effects of physiologic dosages. METHODS: Following ulnar osteotomy, fifty-six rabbits were administered a placebo (nine rabbits), indomethacin (nine rabbits given 12.5 mg/kg daily), or aspirin at various doses and schedules (2.7 mg/kg daily for ten rabbits, 10 mg/kg daily for nine rabbits, 50 mg/kg twice daily for ten rabbits, and 100 mg/kg three times daily for nine rabbits). The aspirin doses were chosen to span the clinical dosing range. The indomethacin group served as a positive control and as a relative comparison with the effect of aspirin. Radiographs were obtained every two weeks and the animals were killed at eight weeks. Mechanical testing was performed on all rabbits except for six selected for histological evaluation. RESULTS: Aspirin delayed bone healing, as demonstrated radiographically and with mechanical testing, in a dose-dependent fashion at salicylate levels equivalent to those resulting from typical human dosing (low-dose aspirin). Receiver operating characteristic analysis demonstrated a plasma salicylate threshold above 20.7 µg/mL predicting delayed bone healing. This approximates a single human dose of 325 mg. Salicylate levels above this threshold were associated with delayed bone healing similar to that caused by indomethacin. Aspirin dosing frequency did not affect bone healing. Mechanical testing was highly predictive of radiographic healing. The interobserver reliability of radiographic assessment of healing at six and eight weeks (kappa = 0.83 and 0.79, respectively) compared favorably with interobserver reliability in previous studies assessing cortical bridging. CONCLUSIONS: In a rabbit ulnar osteotomy model, aspirin delayed bone healing with a threshold equivalent to a human dose of 325 mg.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Bone Regeneration/drug effects , Osteotomy , Ulna/surgery , Wound Healing/drug effects , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Aspirin/administration & dosage , Aspirin/pharmacokinetics , Biomechanical Phenomena , Dose-Response Relationship, Drug , Drug Administration Schedule , Indomethacin/administration & dosage , Indomethacin/pharmacology , Observer Variation , ROC Curve , Rabbits , Radiography , Single-Blind Method , Ulna/diagnostic imaging , Ulna/drug effects , Ulna/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...