Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Infect Immun ; 89(10): e0018021, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34097469

RESUMO

Osteomyelitis can result from the direct inoculation of pathogens into bone during injury or surgery or from spread via the bloodstream, a condition called hematogenous osteomyelitis (HOM). HOM disproportionally affects children, and more than half of cases are caused by Staphylococcus aureus. Laboratory models of osteomyelitis mostly utilize direct injection of bacteria into the bone or implantation of foreign material and therefore do not directly interrogate the pathogenesis of pediatric hematogenous osteomyelitis. In this study, we inoculated mice intravenously and characterized the resultant musculoskeletal infections using two strains isolated from adults (USA300-LAC and NRS384) and five new methicillin-resistant S. aureus isolates from pediatric osteomyelitis patients. All strains were capable of creating stable infections over 5 weeks, although the incidence varied. Micro-computed tomography (microCT) analysis demonstrated decreases in the trabecular bone volume fraction but little effect on bone cortices. Histological assessment revealed differences in the precise focus of musculoskeletal infection, with various mixtures of bone-centered osteomyelitis and joint-centered septic arthritis. Whole-genome sequencing of three new isolates demonstrated distinct strains, two within the USA300 lineage and one USA100 isolate. Interestingly, this USA100 isolate showed a distinct predilection for septic arthritis compared to the other isolates tested, including NRS384 and LAC, which more frequently led to osteomyelitis or mixed bone and joint infections. Collectively, these data outline the feasibility of using pediatric osteomyelitis clinical isolates to study the pathogenesis of HOM in murine models and lay the groundwork for future studies investigating strain-dependent differences in musculoskeletal infection.


Assuntos
Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Osteomielite/microbiologia , Infecções Estafilocócicas/microbiologia , Células 3T3 , Adulto , Animais , Antibacterianos/farmacologia , Artrite Infecciosa/tratamento farmacológico , Artrite Infecciosa/microbiologia , Linhagem Celular , Criança , Humanos , Masculino , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Doenças Musculoesqueléticas/tratamento farmacológico , Doenças Musculoesqueléticas/microbiologia , Osteomielite/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico
2.
Alcohol Clin Exp Res ; 44(6): 1204-1213, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32304578

RESUMO

BACKGROUND: During bone fracture repair, resident mesenchymal stem cells (MSCs) differentiate into chondrocytes, to form a cartilaginous fracture callus, and osteoblasts, to ossify the collagen matrix. Our laboratory previously reported that alcohol administration led to decreased cartilage formation within the fracture callus of rodents and this effect was mitigated by postfracture antioxidant treatment. Forkhead box protein O (FoxO) transcription factors are activated in response to intracellular reactive oxygen species (ROS), and alcohol has been shown to increase ROS. Activation of FoxOs has also been shown to inhibit canonical Wnt signaling, a necessary pathway for MSC differentiation. These findings have led to our hypothesis that alcohol exposure decreases osteochondrogenic differentiation of MSCs through the activation of FoxOs. METHODS: Primary rat MSCs were treated with ethanol (EtOH) and assayed for FoxO expression, FoxO activation, and downstream target expression. Next, MSCs were differentiated toward osteogenic or chondrogenic lineages in the presence of 50 mM EtOH and alterations in osteochondral lineage marker expression were determined. Lastly, osteochondral differentiation experiments were repeated with FoxO1/3 knockdown or with FoxO1/3 inhibitor AS1842856 and osteochondral lineage marker expression was determined. RESULTS: EtOH increased the expression of FoxO3a at mRNA and protein levels in primary cultured MSCs. This was accompanied by an increase in FoxO1 nuclear localization, FoxO1 activation, and downstream catalase expression. Moreover, EtOH exposure decreased expression of osteogenic and chondrogenic lineage markers. FoxO1/3 knockdown restored proosteogenic and prochondrogenic lineage marker expression in the presence of 50 mM EtOH. However, FoxO1/3 inhibitor only restored proosteogenic lineage marker expression. CONCLUSIONS: These data show that EtOH has the ability to inhibit MSC differentiation, and this ability may rely, at least partially, on the activation of FoxO transcription factors.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Proteína Forkhead Box O3/efeitos dos fármacos , Consolidação da Fratura/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Animais , Calo Ósseo/efeitos dos fármacos , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Técnicas de Silenciamento de Genes , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Cultura Primária de Células , Ratos
3.
J Biol Chem ; 290(16): 9959-73, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25713073

RESUMO

Alcohol (EtOH) intoxication is a risk factor for increased morbidity and mortality with traumatic injuries, in part through inhibition of bone fracture healing. Animal models have shown that EtOH decreases fracture callus volume, diameter, and biomechanical strength. Transforming growth factor ß1 (TGF-ß1) and osteopontin (OPN) play important roles in bone remodeling and fracture healing. Mesenchymal stem cells (MSC) reside in bone and are recruited to fracture sites for the healing process. Resident MSC are critical for fracture healing and function as a source of TGF-ß1 induced by local OPN, which acts through the transcription factor myeloid zinc finger 1 (MZF1). The molecular mechanisms responsible for the effect of EtOH on fracture healing are still incompletely understood, and this study investigated the role of EtOH in affecting OPN-dependent TGF-ß1 expression in MSC. We have demonstrated that EtOH inhibits OPN-induced TGF-ß1 protein expression, decreases MZF1-dependent TGF-ß1 transcription and MZF1 transcription, and blocks OPN-induced MZF1 phosphorylation. We also found that PKA signaling enhances OPN-induced TGF-ß1 expression. Last, we showed that EtOH exposure reduces the TGF-ß1 protein levels in mouse fracture callus. We conclude that EtOH acts in a novel mechanism by interfering directly with the OPN-MZF1-TGF-ß1 signaling pathway in MSC.


Assuntos
Etanol/efeitos adversos , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteopontina/farmacologia , Tíbia/efeitos dos fármacos , Fraturas da Tíbia/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Aptâmeros de Nucleotídeos/genética , Aptâmeros de Nucleotídeos/metabolismo , Diferenciação Celular , Consolidação da Fratura/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Luciferases/genética , Luciferases/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Osteopontina/metabolismo , Fosforilação , Transdução de Sinais , Tíbia/lesões , Tíbia/metabolismo , Fraturas da Tíbia/genética , Fraturas da Tíbia/patologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
4.
Alcohol Clin Exp Res ; 36(12): 2095-103, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22691115

RESUMO

BACKGROUND: Alcohol abuse is a risk factor for bone damage and fracture-related complications. Through precise ß-catenin signaling, canonical Wnt signaling plays a key role in fracture repair by promoting the differentiation of new bone and cartilage cells. In this study, we examined the effects of alcohol on the Wnt pathway in injured bone using a murine model of alcohol-induced impaired fracture healing. METHODS: Male C57Bl/6 or T cell factor (TCF)-transgenic mice were administered 3 daily intraperitoneal doses of alcohol or saline. One hour following the final injection, mice were subjected to a stabilized, mid-shaft tibial fracture. Injured and contralateral tibias were harvested at 6, 9, or 14 days post-fracture for the analysis of biomechanical strength, callus tissue composition, and Wnt/ß-catenin signaling. RESULTS: Acute alcohol treatment was associated with a significant decrease in fracture callus volume, diameter, and biomechanical strength at day 14 post-fracture. Histology revealed an alcohol-related reduction in cartilage and bone formation at the fracture site, and that alcohol inhibited normal cartilage maturation. Acute alcohol exposure caused a significant 2.3-fold increase in total ß-catenin protein at day 6 and a significant decrease of 53 and 56% at days 9 and 14, respectively. lacZ staining in ß-galactosidase-expressing TCF-transgenic mice revealed spatial and quantitative differences in Wnt-specific transcriptional activation at day 6 in the alcohol group. Days 9 and 14 post-fracture showed that acute alcohol exposure decreased Wnt transcriptional activation, which correlates with the modulation of total ß-catenin protein levels observed at these time points. CONCLUSIONS: Acute alcohol exposure resulted in significant impairment of fracture callus tissue formation, perturbation of the key Wnt pathway protein ß-catenin, and disruption of normal Wnt-mediated transcription. These data suggest that the canonical Wnt pathway is a target for alcohol in bone and may partially explain why impaired fracture healing is observed in alcohol-abusing individuals.


Assuntos
Calo Ósseo/efeitos dos fármacos , Etanol/efeitos adversos , Consolidação da Fratura/efeitos dos fármacos , beta Catenina/antagonistas & inibidores , Animais , Calo Ósseo/química , Calo Ósseo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fraturas da Tíbia/fisiopatologia , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/análise , beta Catenina/efeitos dos fármacos
5.
Bone ; 148: 115941, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33813068

RESUMO

Mitochondria are essential organelles that form highly complex, interconnected dynamic networks inside cells. The GTPase mitofusin 2 (MFN2) is a highly conserved outer mitochondrial membrane protein involved in the regulation of mitochondrial morphology, which can affect various metabolic and signaling functions. The role of mitochondria in bone formation remains unclear. Since MFN2 levels increase during osteoblast (OB) differentiation, we investigated the role of MFN2 in the osteolineage by crossing mice bearing floxed Mfn2 alleles with those bearing Prx-cre to generate cohorts of conditional knock out (cKO) animals. By ex vivo microCT, cKO female mice, but not males, display an increase in cortical thickness at 8, 18, and 30 weeks, compared to wild-type (WT) littermate controls. However, the cortical anabolic response to mechanical loading was not different between genotypes. To address how Mfn2 deficiency affects OB differentiation, bone marrow-derived mesenchymal stromal cells (MSCs) from both wild-type and cKO mice were cultured in osteogenic media with different levels of ß-glycerophosphate. cKO MSCs show increased mineralization and expression of multiple markers of OB differentiation only at the lower dose. Interestingly, despite showing the expected mitochondrial rounding and fragmentation due to loss of MFN2, cKO MSCs have an increase in oxygen consumption during the first 7 days of OB differentiation. Thus, in the early phases of osteogenesis, MFN2 restrains oxygen consumption thereby limiting differentiation and cortical bone accrual during homeostasis in vivo.


Assuntos
GTP Fosfo-Hidrolases , Osteogênese , Animais , Diferenciação Celular , Osso Cortical/diagnóstico por imagem , Feminino , GTP Fosfo-Hidrolases/genética , Camundongos , Camundongos Knockout
6.
Cells ; 9(10)2020 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-32987689

RESUMO

Bone infections, also known as infectious osteomyelitis, are accompanied by significant inflammation, osteolysis, and necrosis. Osteoclasts (OCs) are the bone-resorbing cells that work in concert with osteoblasts and osteocytes to properly maintain skeletal health and are well known to respond to inflammation by increasing their resorptive activity. OCs have typically been viewed merely as effectors of pathologic bone resorption, but recent evidence suggests they may play an active role in the progression of infections through direct effects on pathogens and via the immune system. This review discusses the host- and pathogen-derived factors involved in the in generation of OCs during infection, the crosstalk between OCs and immune cells, and the role of OC lineage cells in the growth and survival of pathogens, and highlights unanswered questions in the field.


Assuntos
Bactérias/metabolismo , Reabsorção Óssea/imunologia , Reabsorção Óssea/microbiologia , Osso e Ossos/imunologia , Osso e Ossos/microbiologia , Linhagem da Célula , Imunomodulação , Osteoclastos/patologia , Animais , Osso e Ossos/patologia , Humanos
7.
mBio ; 10(5)2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31615966

RESUMO

Osteomyelitis (OM), or inflammation of bone tissue, occurs most frequently as a result of bacterial infection and severely perturbs bone structure. OM is predominantly caused by Staphylococcus aureus, and even with proper treatment, OM has a high rate of recurrence and chronicity. While S. aureus has been shown to infect osteoblasts, it remains unclear whether osteoclasts (OCs) are also a target of intracellular infection. Here, we demonstrate the ability of S. aureus to intracellularly infect and divide within OCs. OCs were differentiated from bone marrow macrophages (BMMs) by exposure to receptor activator of nuclear factor kappa-B ligand (RANKL). By utilizing an intracellular survival assay and flow cytometry, we found that at 18 h postinfection the intracellular burden of S. aureus increased dramatically in cells with at least 2 days of RANKL exposure, while the bacterial burden decreased in BMMs. To further explore the signals downstream of RANKL, we manipulated factors controlling OC differentiation, NFATc1 and alternative NF-κB, and found that intracellular bacterial growth correlates with NFATc1 levels in RANKL-treated cells. Confocal and time-lapse microscopy in mature OCs showed a range of intracellular infection that correlated inversely with S. aureus-phagolysosome colocalization. The propensity of OCs to become infected, paired with their diminished bactericidal capacity compared to BMMs, could promote OM progression by allowing S. aureus to evade initial immune regulation and proliferate at the periphery of lesions where OCs are most abundant.IMPORTANCE The inflammation of bone tissue is called osteomyelitis, and most cases are caused by an infection with the bacterium Staphylococcus aureus To date, the bone-building cells, osteoblasts, have been implicated in the progression of these infections, but not much is known about how the bone-resorbing cells, osteoclasts, participate. In this study, we show that S. aureus can infect osteoclasts and proliferate inside these cells, whereas bone-residing macrophages, immune cells related to osteoclasts, destroy the bacteria. These findings elucidate a unique role for osteoclasts to harbor bacteria during infection, providing a possible mechanism by which bacteria could evade destruction by the immune system.


Assuntos
Osteoclastos/microbiologia , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidade , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Diferenciação Celular , Células Cultivadas , Feminino , Macrófagos/metabolismo , Masculino , Camundongos , Osteoblastos/microbiologia , Osteomielite/metabolismo , Osteomielite/microbiologia , Fagossomos/metabolismo , Ligante RANK/metabolismo , Staphylococcus aureus/efeitos dos fármacos
8.
Alcohol ; 77: 11-18, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30763905

RESUMO

On January 26, 2018, the 23rd annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held at the University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado. The meeting consisted of plenary sessions with oral presentations and a poster presentation session. There were four plenary sessions that covered a wide range of topics relating to alcohol use: Alcohol and Liver Disease; Alcohol, Inflammation and Immune Response; Alcohol and Organ Injury; Heath Consequences and Alcohol Drinking. The meeting provided a forum for the presentation and discussion of novel research findings regarding alcohol use and immunology.


Assuntos
Consumo de Bebidas Alcoólicas/imunologia , Alcoolismo/imunologia , Pesquisa Biomédica/tendências , Congressos como Assunto/tendências , Imunidade Celular/imunologia , Consumo de Bebidas Alcoólicas/patologia , Alcoolismo/diagnóstico , Animais , Pesquisa Biomédica/métodos , Colorado , Humanos , Imunidade Celular/efeitos dos fármacos
9.
J Orthop Trauma ; 32(6): 288-295, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29672339

RESUMO

OBJECTIVES: To explore how alcohol affects the BMP-2 signaling pathway, which is known to play a critical role in bone and cartilage formation during fracture healing. METHODS: A rat model was used to demonstrate the detrimental effects of alcohol exposure on tibia fracture healing. Specific components of the BMP-2 pathway were analyzed in fracture callus on days 3, 7, 14, and 21 after fracture via western immunoassays and enzyme-linked immunosorbent assay. RESULTS: Alcohol exposure before tibia fracture demonstrated attenuation of downstream BMP-2 signaling. The BMP-2 antagonist, Chordin, may be the central component of the BMP-2-related changes demonstrated in this study. Although alcohol affected BMP-related proteins at all time points, it seems that day 14 after fracture is a critical time point for alcohol-related modulation of callus formation in our model. CONCLUSIONS: This study may provide the scientific basis for further studies addressing whether the application of exogenous BMP-2 in patients with a history of alcohol abuse who sustain long bone fractures may or may not be of benefit.


Assuntos
Consumo de Bebidas Alcoólicas , Proteína Morfogenética Óssea 2/metabolismo , Etanol/farmacologia , Consolidação da Fratura/efeitos dos fármacos , Fraturas da Tíbia/metabolismo , Animais , Modelos Animais de Doenças , Ratos Sprague-Dawley , Fraturas da Tíbia/tratamento farmacológico
10.
J Orthop Surg Res ; 13(1): 101, 2018 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-29699560

RESUMO

BACKGROUND: Alcohol consumption is a risk factor for impaired fracture healing, though the mechanism(s) by which this occurs are not well understood. Our laboratory has previously shown that episodic alcohol exposure of rodents negatively affects fracture callus development, callus biomechanics, and cellular signaling which regulates stem cell differentiation. Here, we examine whether alcohol alters chemokine expression and/or signaling activity in the mouse fracture callus during early fracture healing. METHODS: A mouse model for alcohol-impaired tibia fracture healing was utilized. Early fracture callus was examined for alcohol-effects on tissue composition, expression of chemokines involved in MSC migration to the fracture site, and biomechanics. The effects of alcohol on MSC migration and cell adhesion receptors were examined in an in vitro system. RESULTS: Mice exposed to alcohol showed decreased evidence of external callus formation, decreased callus-related osteopontin (OPN) expression levels, and decreased biomechanical stiffness. Alcohol exposure decreased rOPN-mediated MSC migration and integrin ß1 receptor expression in vitro. CONCLUSIONS: The effects of alcohol exposure demonstrated here on fracture callus-associated OPN expression, rOPN-mediated MSC migration in vitro, and MSC integrin ß1 receptor expression in vitro have not been previously reported. Understanding the effects of alcohol exposure on the early stages of fracture repair may allow timely initiation of treatment to mitigate the long-term complications of delayed healing and/or fracture non-union.


Assuntos
Movimento Celular/efeitos dos fármacos , Etanol/toxicidade , Consolidação da Fratura/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteopontina/antagonistas & inibidores , Osteopontina/biossíntese , Animais , Movimento Celular/fisiologia , Consolidação da Fratura/fisiologia , Expressão Gênica , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteopontina/genética , Tíbia/efeitos dos fármacos , Tíbia/lesões , Tíbia/metabolismo
11.
J Orthop Res ; 34(12): 2106-2115, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26998841

RESUMO

The process of fracture healing is complex, and poor or incomplete healing remains a significant health problem. Proper fracture healing relies upon resident mesenchymal stem cell (MSC) differentiation into chondrocytes and osteoblasts, which are necessary for callus formation and ossification. Alcohol abuse is a leading contributor to poor fracture healing. Although the mechanism behind this action is unknown, excessive alcohol consumption is known to promote systemic oxidative stress. The family of FoxO transcription factors is activated by oxidative stress, and FoxO activation antagonizes Wnt signaling, which regulates mesenchymal stem cell differentiation. We hypothesize that alcohol exposure increases oxidative stress leading to deficient fracture repair by activating FoxO transcription factors within the fracture callus which disrupts chondrogenesis of mesenchymal stem cells. Our laboratory has developed an experimental model of delayed fracture union in mice using ethanol administration. We have found that ethanol administration significantly decreases external, cartilaginous callus formation, and hallmarks of endochondral ossification, and these changes are concomitant with increases in FoxO expression and markers of activation in fracture callus tissue of these mice. We were able to prevent these alcohol-induced effects with the administration of the antioxidant n-acetyl cysteine (NAC), suggesting that alcohol-induced oxidative stress produces the perturbed endochondral ossification and FoxO expression. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:2106-2115, 2016.


Assuntos
Calo Ósseo/efeitos dos fármacos , Depressores do Sistema Nervoso Central/efeitos adversos , Etanol/efeitos adversos , Fatores de Transcrição Forkhead/metabolismo , Consolidação da Fratura/efeitos dos fármacos , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Animais , Calo Ósseo/metabolismo , Condrogênese/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Sequestradores de Radicais Livres/farmacologia , Sequestradores de Radicais Livres/uso terapêutico , Masculino , Camundongos Endogâmicos C57BL , Distribuição Aleatória
12.
J Orthop Trauma ; 25(8): 516-21, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21738068

RESUMO

OBJECTIVES: Alcohol consumption is a known risk factor for traumatic injuries of all types and has been shown to produce detrimental effects on bone metabolism. Although the mechanisms responsible for these detrimental effects are not well characterized, oxidative stress from alcohol exposure appears to play a central role. This study was designed to examine the effect of a short-term binge alcohol consumption pattern on fracture repair and the effect of an antioxidant, N-acetylcysteine, on fracture healing after binge alcohol consumption. METHODS: One hundred forty-four adult male Sprague-Dawley rats underwent unilateral closed femur fracture after injection of either saline or alcohol to simulate a binge alcohol cycle. Animals in the antioxidant treatment group received daily N-acetylcysteine after fracture. Femurs were harvested at 1, 2, 4, and 6 weeks after injury and underwent biomechanical testing and histologic analysis. RESULTS: Binge alcohol administration was associated with significant decreases in biomechanical strength at 1- and 2-week time points with a trend toward decreased strength at 4- and 6-week time points as well. Alcohol-treated animals had less cartilage component within the fracture callus and healed primarily by intramembranous ossification. Administration of N-acetylcysteine in alcohol-treated animals improved biomechanical strength to levels comparable to the control animals and was associated with increased endochondral ossification. CONCLUSIONS: Our results indicate that binge alcohol alters the quality of fracture healing after a traumatic injury and that concurrent administration of an antioxidant is able to reverse these effects.


Assuntos
Alcoolismo/fisiopatologia , Antioxidantes/administração & dosagem , Etanol/toxicidade , Fraturas do Fêmur/tratamento farmacológico , Fraturas do Fêmur/fisiopatologia , Consolidação da Fratura/efeitos dos fármacos , Consolidação da Fratura/fisiologia , Alcoolismo/complicações , Animais , Relação Dose-Resposta a Droga , Fraturas do Fêmur/complicações , Masculino , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA