Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Am Acad Orthop Surg ; 23(1): 1-6, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25538125

RESUMO

Patients who sustain orthopaedic trauma are at risk for developing deep venous thrombosis and symptomatic pulmonary emboli. The prevention of venous thromboembolism has moved to the forefront of patient safety initiatives, resulting in the formation of various guidelines to assist the practitioner. Recommendations for venous thromboembolism prophylaxis in the orthopaedic trauma patient exist, but there is insufficient evidence in the literature to make strong recommendations regarding type and duration of prophylaxis. The associated morbidity of chemical anticoagulants used in the orthopaedic trauma patient must also be taken into consideration, specifically the increased risk of bleeding. Mechanical prophylaxis is used in place of, or in addition to, these medications in certain situations. New, potentially superior anticoagulants have been developed but are still understudied. Larger studies are needed to further define the type and duration of deep venous thrombosis prophylaxis in the orthopaedic trauma patient.


Assuntos
Extremidade Inferior/lesões , Procedimentos Ortopédicos , Extremidade Superior/lesões , Tromboembolia Venosa/prevenção & controle , Humanos , Complicações Pós-Operatórias/prevenção & controle , Fatores de Risco , Tromboembolia Venosa/epidemiologia , Tromboembolia Venosa/fisiopatologia
2.
J Bone Miner Res ; 29(6): 1478-91, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24677136

RESUMO

Previous studies showed that loss of tumor necrosis factor α (TNFα) signaling delayed fracture healing by delaying chondrocyte apoptosis and cartilage resorption. Mechanistic studies showed that TNFα induced Fas expression within chondrocytes; however, the degree to which chondrocyte apoptosis is mediated by TNFα alone or dependent on the induction of Fas is unclear. This question was addressed by assessing fracture healing in Fas-deficient B6.MRL/Fas(lpr) /J mice. Loss of Fas delayed cartilage resorption but also lowered bone fraction in the calluses. The reduced bone fraction was related to elevated rates of coupled bone turnover in the B6.MRL/Fas(lpr) /J calluses, as evidenced by higher osteoclast numbers and increased osteogenesis. Analysis of the apoptotic marker caspase 3 showed fewer positive chondrocytes and osteoclasts in calluses of B6.MRL/Fas(lpr) /J mice. To determine if an active autoimmune state contributed to increased bone turnover, the levels of activated T cells and Treg cells were assessed. B6.MRL/Fas(lpr) /J mice had elevated Treg cells in both spleens and bones of B6.MRL/Fas(lpr) /J but decreased percentage of activated T cells in bone tissues. Fracture led to ∼30% to 60% systemic increase in Treg cells in both wild-type and B6.MRL/Fas(lpr) /J bone tissues during the period of cartilage formation and resorption but either decreased (wild type) or left unchanged (B6.MRL/Fas(lpr) /J) the numbers of activated T cells in bone. These results show that an active autoimmune state is inhibited during the period of cartilage resorption and suggest that iTreg cells play a functional role in this process. These data show that loss of Fas activity specifically in chondrocytes prolonged the life span of chondrocytes and that Fas synergized with TNFα signaling to mediate chondrocyte apoptosis. Conversely, loss of Fas systemically led to increased osteoclast numbers during later periods of fracture healing and increased osteogenesis. These findings suggest that retention of viable chondrocytes locally inhibits osteoclast activity or matrix proteolysis during cartilage resorption.


Assuntos
Consolidação da Fratura , Fraturas Ósseas/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Linfócitos T Reguladores/imunologia , Receptor fas/deficiência , Animais , Apoptose , Fenômenos Biomecânicos , Remodelação Óssea/genética , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Calo Ósseo/diagnóstico por imagem , Calo Ósseo/patologia , Cartilagem/metabolismo , Cartilagem/patologia , Modelos Animais de Doenças , Fraturas Ósseas/diagnóstico por imagem , Fraturas Ósseas/patologia , Fraturas Ósseas/fisiopatologia , Inflamação/patologia , Lúpus Eritematoso Sistêmico/diagnóstico por imagem , Lúpus Eritematoso Sistêmico/patologia , Lúpus Eritematoso Sistêmico/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Especificidade de Órgãos/genética , Osteoclastos/patologia , Osteogênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Microtomografia por Raio-X , Receptor fas/metabolismo
3.
J Cell Physiol ; 228(11): 2232-42, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23625810

RESUMO

Runx2 and Runx3 are known to be expressed in the growth plate during endochondral bone formation. Here we addressed the functional role of Runx3 as distinct from Runx2 by using two models of postnatal bone repair: fracture healing that proceeds by an endochondral process and marrow ablation that proceeds by only an intramembranous process. Both Runx2 and Runx3 mRNAs were differentially up regulated during fracture healing. In contrast, only Runx2 showed increased expression after marrow ablation. During fracture healing, Runx3 was expressed earlier than Runx2, was concurrent with the period of chondrogenesis, and coincident with maximal aggrecan expression a protein associated with proliferating and permanent cartilage. Immunohistological analysis showed Runx3 protein was also expressed by chondrocytes in vivo. In contrast, Runx2 was expressed later during chondrocyte hypertrophy, and primary bone formation. The functional activities of Runx3 during chondrocyte differentiation were assessed by examining its regulatory actions on aggrecan gene expression. Aggrecan mRNA levels and aggrecan promoter activity were enhanced in response to the over-expression of either Runx2 and Runx3 in ATDC5 chondrogenic cell line, while sh-RNA knocked down of each Runx protein showed that only Runx3 knock down specifically suppressed aggrecan mRNA expression and promoter activity. ChIP assay demonstrated that Runx3 interactions were selective to sites within the aggrecan promoter and were only observed during early periods of chondrogenesis before hypertrophy. Our studies suggest that Runx3 positively regulates aggrecan expression and suggest that its function is more limited to cartilage development than to bone. In aggregate these data further suggest that the various members of the Runx transcription factors are involved in the coordination of chondrocyte development, maturation, and hypertrophy during endochondral bone formation.


Assuntos
Agrecanas/genética , Cartilagem/crescimento & desenvolvimento , Cartilagem/metabolismo , Condrogênese/genética , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica , Agrecanas/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Especificidade de Órgãos/genética , Osteogênese/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Tempo
4.
J Biol Chem ; 287(19): 15718-27, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22403399

RESUMO

The differentiation of osteoblasts from their precursors, mesenchymal stem cells, is an important component of bone homeostasis as well as fracture healing. The A2B adenosine receptor (A2BAR) is a Gα(s)/α(q)-protein-coupled receptor that signals via cAMP. cAMP-mediated signaling has been demonstrated to regulate the differentiation of mesenchymal stem cells (MSCs) into various skeletal tissue lineages. Here, we studied the role of this receptor in the differentiation of MSCs to osteoblasts. In vitro differentiation of bone marrow-derived MSCs from A2BAR KO mice resulted in lower expression of osteoblast differentiation transcription factors and the development of fewer mineralized nodules, as compared with WT mice. The mechanism of effect involves, at least partially, cAMP as indicated by experiments involving activation of the A2BAR or addition of a cAMP analog during differentiation. Intriguingly, in vivo, microcomputed tomography analysis of adult femurs showed lower bone density in A2BAR KO mice as compared with WT. Furthermore, A2BAR KO mice display a delay in normal fracture physiology with lower expression of osteoblast differentiation genes. Thus, our study identified the A2BAR as a new regulator of osteoblast differentiation, bone formation, and fracture repair.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Osteogênese/fisiologia , Receptor A2B de Adenosina/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Densidade Óssea , Calcificação Fisiológica/efeitos dos fármacos , Calcificação Fisiológica/genética , Calcificação Fisiológica/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Feminino , Fêmur/metabolismo , Consolidação da Fratura/efeitos dos fármacos , Consolidação da Fratura/genética , Consolidação da Fratura/fisiologia , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/citologia , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Receptor A2B de Adenosina/genética , Fator de Transcrição Sp7 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Microtomografia por Raio-X
5.
Injury ; 43(3): 274-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21689817

RESUMO

Whilst the majority of fractures heal normally, it is estimated that ∼10% of fractures exhibit some level of delayed or impaired healing. Although radiography is the primary diagnostic tool to assess the progression of fracture healing, radiographic features only qualitatively correlate with tissue level increases in mineral content and do not quantitatively measure underlying biological processes that are associated with the progression of healing. Specific metaloproteinases have been shown to be essential to processes of both angiogenesis and mineralised cartilage resorption and bone remodelling at different phases of fracture healing. The aim of this study was to determine the potential of using a simple urine based assay of the activity of two MMPs as a means of assessing the biological progression of fracture healing through the endochondral phase of healing. Using a standard mid-diaphyseal murine model of femoral fracture, MMP9 and MMP13 proteins and enzymatic activity levels were quantified in the urine of mice across the time-course of fracture healing and compared to the mRNA and protein expression profiles in the calluses. Both urinary MMP9 and MMP13 protein and enzymatic activity levels, assessed by Western blot, zymogram and specific MMP fluorometric substrate assays, corresponded to mRNA expression and immunohistologic assays of the proteins within callus tissues. These studies suggest that urinary levels of MMP9 and MMP13 may have potential as metabolic markers to monitor the progression of fracture healing.


Assuntos
Remodelação Óssea , Fraturas do Fêmur/urina , Consolidação da Fratura , Metaloproteinase 13 da Matriz/urina , Metaloproteinase 9 da Matriz/urina , RNA Mensageiro/urina , Animais , Biomarcadores/urina , Western Blotting , Fraturas do Fêmur/fisiopatologia , Masculino , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Camundongos , Valor Preditivo dos Testes
6.
J Trauma ; 70(4): 948-53, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20693926

RESUMO

BACKGROUND: The acute-phase response (APR) is critical to the body's ability to successfully respond to injury. A murine model of closed unilateral femur fractures and bilateral femur fracture were used to study the effect of injury magnitude on this response. METHODS: Standardized unilateral femur fracture and bilateral femur fracture in mice were performed. The femur fracture sites, livers, and serum were harvested over time after injury. Changes in mRNA expression of cytokines, hepatic acute-phase proteins, and serum cytokines overtime were measured. RESULTS: There was a rapid and short-lived hepatic APR to fracture injuries. The overall pattern in both models was similar. Both acute-phase proteins' mRNA (fibrinogen-γ and serum amyloid A-3) showed increased mRNA expression over baseline within the first 48 hours and their levels positively correlated with the extent of injury. However, increased severity of injury resulted in a delayed induction of the APR. A similar effect on the gene expression of cytokines (interleukin [IL]-1ß, IL-6, and tumor necrosis factor-α) at the fracture site was seen. Serum IL-6 levels increased with increased injury and showed no delay between injury models. CONCLUSIONS: Greater severity of injury resulted in a delayed induction of the liver's APR and a diminished expression of cytokines at the fracture site. Serum IL-6 levels were calibrated to the extent of the injury, and changes may represent mechanisms by which the local organ responses to injury are regulated by the injury magnitude.


Assuntos
Proteínas de Fase Aguda/genética , Reação de Fase Aguda/genética , Fraturas do Fêmur/genética , Expressão Gênica , Fígado/metabolismo , RNA Mensageiro/genética , Proteínas de Fase Aguda/biossíntese , Reação de Fase Aguda/metabolismo , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Fraturas do Fêmur/metabolismo , Interleucina-6/biossíntese , Interleucina-6/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
7.
Arthritis Rheum ; 62(4): 1108-18, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20131271

RESUMO

OBJECTIVE: To characterize patterns of molecular expression that lead to cartilage formation in vivo in a postnatal setting, by profiling messenger RNA expression across the time course of mechanically induced chondrogenesis. METHODS: Retired breeder Sprague-Dawley rats underwent a noncritical-sized transverse femoral osteotomy. Experimental animals (n = 45) were subjected to bending stimulation (60 degrees cyclic motion in the sagittal plane for 15 minutes/day) of the osteotomy gap beginning on day 10 after the operation. Control animals (n = 32) experienced continuous rigid fixation. Messenger RNA isolated on days 10, 17, 24, and 38 after surgery was analyzed using a microarray containing 608 genes involved in skeletal development, tissue differentiation, fracture healing, and mechanotransduction. The glycosaminoglycan (GAG) content in the stimulated tissues was compared with that in native articular cartilage as a means of assessing the progression of chondrogenic development of the tissues. RESULTS: The majority of the 100 genes that were differentially expressed were up-regulated in response to mechanical stimulation. Many of these genes are associated with articular cartilage development and maintenance, diarthrodial joint development, cell adhesion, extracellular matrix synthesis, signal transduction, and skeletal development. Quantitative real-time polymerase chain reaction results were consistent with the microarray findings. The GAG content of the stimulated tissues increased over time and was no different from that of articular cartilage on day 38 after surgery. CONCLUSION: Our findings indicate that mechanical stimulation causes up-regulation of genes that are principally involved in joint cavity morphogenesis and critical to articular cartilage function. Further study of this type of stimulation may identify key signaling events required for postnatal hyaline cartilage formation.


Assuntos
Cartilagem Articular/fisiologia , Perfilação da Expressão Gênica , Transcrição Gênica , Animais , Cartilagem Articular/metabolismo , Condrogênese/genética , Condrogênese/fisiologia , Fêmur/cirurgia , Regulação da Expressão Gênica , Masculino , Modelos Animais , Família Multigênica/genética , Análise de Sequência com Séries de Oligonucleotídeos , Osteotomia , Postura , RNA Mensageiro/genética , RNA Mensageiro/isolamento & purificação , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
J Bone Miner Res ; 25(4): 724-33, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19839770

RESUMO

Hypophosphatemia leads to rickets and osteomalacia, the latter of which results in decreased biomechanical integrity of bones, accompanied by poor fracture healing. Impaired phosphate-dependent apoptosis of hypertrophic chondrocytes is the molecular basis for rickets. However, the underlying pathophysiology of impaired fracture healing has not been characterized previously. To address the role of phosphate in fracture repair, mice were placed on a phosphate-restricted diet 2 days prior to or 3 days after induction of a mid-diaphyseal femoral fracture to assess the effects of phosphate deficiency on the initial recruitment of mesenchymal stem cells and their subsequent differentiation. Histologic and micro-computed tomographic (microCT) analyses demonstrated that both phosphate restriction models dramatically impaired fracture healing primarily owing to a defect in differentiation along the chondrogenic lineage. Based on Sox9 and Sox5 mRNA levels, neither the initial recruitment of cells to the callus nor their lineage commitment was effected by hypophosphatemia. However, differentiation of these cells was impaired in association with impaired bone morphogenetic protein (BMP) signaling. In vivo ectopic bone-formation assays and in vitro investigations in ST2 stromal cells confirmed that phosphate restriction leads to BMP-2 resistance. Marrow ablation studies demonstrate that hypophosphatemia has different effects on injury-induced intramembranous bone formation compared with endochondral bone formation. Thus phosphate plays an important role in the skeleton that extends beyond mineralized matrix formation and growth plate maturation and is critical for endochondral bone repair.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Fêmur/lesões , Consolidação da Fratura , Fraturas Ósseas/metabolismo , Fosfatos/metabolismo , Animais , Calo Ósseo/metabolismo , Diferenciação Celular , Movimento Celular , Dieta , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise , Fatores de Transcrição SOX9/análise , Fatores de Transcrição SOXD/análise
9.
Bone ; 43(6): 1031-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18761113

RESUMO

Fracture healing involves multiple stages of repair and coordinated actions of multiple cell types. Consequently, it may be possible to enhance healing through treatment strategies that target more than one repair process or cell type. The goal of this study was to determine the combined effects of recombinant human bone morphogenetic protein 7 (rhBMP-7) and parathyroid hormone (PTH(1-34)) on metaphyseal bone healing. A wedge-shaped defect was created in the lateral aspect of the distal tibia in female New Zealand white rabbits (n=64) and was filled with tricalcium phosphate (TCP). Animals were assigned to four groups: 1) BMP-7 and PTH; 2) BMP-7; 3) PTH; and 4) control (TCP alone). In groups 1 and 2, 200 microg rhBMP-7 was incorporated into the TCP. Animals received daily subcutaneous injections of 10 microg/kg PTH(1-34) (groups 1 and 3) or saline (groups 2 and 4). Healing at 4 weeks was assessed using micro-computed tomography, histology, immunohistochemistry, and mechanical testing. Combined treatment with rhBMP-7 and PTH resulted in increased callus total volume (TV), mineralized volume (BV), average cross-sectional area, and bone mineral content (BMC) as compared to the control group (p<0.02). BV and BMC were also higher in the combined treatment group as compared to the BMP-7 group (p<0.02); however, tissue mineral density was highest in the BMP-7 group (p=0.002). New bone formation in the BMP-7 group was largely restricted to the defect site, while PTH promoted bone formation throughout the defect and surrounding regions. Combined treatment led to greater quantities of woven trabecular bone, increased trabecular thickness, decreased trabecular separation (p<0.04), and a trend towards increased numbers of osteoclasts (p=0.09). Combined treatment also resulted in increased torsional rigidity and compressive strength as compared to the control and BMP-7 groups (p<0.001). These results suggest that the improvements in mechanical function obtained with the combined treatment resulted from differing biological activities of rhBMP-7 and PTH. While the activities of rhBMP-7 appeared to be strictly anabolic, those of PTH appeared to work in the context of coupled remodeling. The combination of both agents led to greater bone volume as well as better microstructural organization and integration of this bone with the surrounding tissues.


Assuntos
Proteína Morfogenética Óssea 7/farmacologia , Consolidação da Fratura/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Animais , Fenômenos Biomecânicos , Densidade Óssea , Feminino , Humanos , Imuno-Histoquímica , Coelhos , Proteínas Recombinantes/farmacologia , Tomografia Computadorizada por Raios X
10.
J Bone Miner Res ; 22(12): 1903-12, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17680724

RESUMO

UNLABELLED: Studies have shown that systemic PTH treatment enhanced the rate of bone repair in rodent models. However, the mechanisms through which PTH affects bone repair have not been elucidated. In these studies we show that PTH primarily enhanced the earliest stages of endochondral bone repair by increasing chondrocyte recruitment and rate of differentiation. In coordination with these cellular events, we observed an increased level of canonical Wnt-signaling in PTH-treated bones at multiple time-points across the time-course of fracture repair, supporting the conclusion that PTH responses are at least in part mediated through Wnt signaling. INTRODUCTION: Since FDA approval of PTH [PTH(1-34); Forteo] as a treatment for osteoporosis, there has been interest in its use in other musculoskeletal conditions. Fracture repair is one area in which PTH may have a significant clinical impact. Multiple animal studies have shown that systemic PTH treatment of healing fractures increased both callus volume and return of mechanical competence in models of fracture healing. Whereas the potential for PTH has been established, the mechanism(s) by which PTH produces these effects remain elusive. MATERIALS AND METHODS: Closed femoral fractures were generated in 8-wk-old male C57Bl/6 mice followed by daily systemic injections of either saline (control) or 30 microg/kg PTH(1-34) for 14 days after fracture. Bones were harvested at days 2, 3, 5, 7, 10, 14, 21, and 28 after fracture and analyzed at the tissue level by radiography and histomorphometry and at the molecular and biochemical levels level by RNase protection assay (RPA), real-time PCR, and Western blot analysis. RESULTS: Quantitative muCT analysis showed that PTH treatment induced a larger callus cross-sectional area, length, and total volume compared with controls. Molecular analysis of the expression of extracellular matrix genes associated with chondrogenesis and osteogenesis showed that PTH treated fractures displayed a 3-fold greater increase in chondrogenesis relative to osteogenesis over the course of the repair process. In addition, chondrocyte hypertrophy occurred earlier in the PTH-treated callus tissues. Analysis of the expression of potential mediators of PTH actions showed that PTH treatment significantly induced the expression of Wnts 4, 5a, 5b, and 10b and increased levels of unphosphorylated, nuclear localized beta-catenin protein, a central feature of canonical Wnt signaling. CONCLUSIONS: These results showed that the PTH-mediated enhancement of fracture repair is primarily associated with an amplification of chondrocyte recruitment and maturation in the early fracture callus. Associated with these cellular effects, we observed an increase in canonical Wnt signaling supporting the conclusion that PTH effects on bone repair are mediated at least in part through the activation of Wnt-signaling pathways.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Condrogênese/efeitos dos fármacos , Fraturas do Fêmur/tratamento farmacológico , Consolidação da Fratura/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Teriparatida/farmacologia , Proteínas Wnt/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Modelos Animais de Doenças , Fraturas do Fêmur/metabolismo , Fraturas do Fêmur/patologia , Masculino , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA