Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 11(2): e0147465, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26849055

RESUMO

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


Assuntos
Consolidação da Fratura/genética , Consolidação da Fratura/imunologia , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Animais , Fenômenos Biomecânicos , Reabsorção Óssea/genética , Reabsorção Óssea/imunologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/genética , Osteogênese/imunologia , Fenótipo , Microtomografia por Raio-X
2.
J Orthop Res ; 32(12): 1589-95, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25183200

RESUMO

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


Assuntos
Calo Ósseo/fisiologia , Consolidação da Fratura/fisiologia , Animais , Fenômenos Biomecânicos , Masculino , Ratos Wistar , Fatores de Tempo
3.
PLoS One ; 8(11): e81341, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260573

RESUMO

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


Assuntos
Calo Ósseo/imunologia , Complemento C3a/genética , Complemento C5a/genética , Consolidação da Fratura/genética , Fraturas Ósseas/genética , Animais , Fenômenos Biomecânicos , Calo Ósseo/diagnóstico por imagem , Calo Ósseo/patologia , Ativação do Complemento/genética , Complemento C3a/deficiência , Complemento C3a/imunologia , Complemento C5a/deficiência , Complemento C5a/imunologia , Elasticidade , Fêmur/diagnóstico por imagem , Fêmur/lesões , Consolidação da Fratura/imunologia , Fraturas Ósseas/diagnóstico por imagem , Fraturas Ósseas/imunologia , Deleção de Genes , Expressão Gênica/imunologia , Dureza , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Masculino , Camundongos , Microtomografia por Raio-X
4.
J Mater Sci Mater Med ; 24(10): 2417-27, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23801500

RESUMO

Hydrogel coatings prepared from reactive star shaped polyethylene oxide based prepolymers (NCO-sP(EO-stat-PO)) minimize unspecific protein adsorption in vitro, while proteins immobilized on NCO-sP(EO-stat-PO) coatings retain their structure and biological function. The aim of the present study was to assess biocompatibility and the effect on early osseointegrative properties of a NCO-sP(EO-stat-PO) coating with additional RGD-peptides and augmentation with bone morphogenetic protein-4 (BMP) used on a medical grade high-density polyethylene (HDPE) base under in vivo circumstances. For testing of biocompatibility dishes with large amounts of bulk NCO-sP(EO-stat-PO) were implanted subcutaneously into 14 Wistar rats. In a second set-up functionalization of implants with ultrathin surface layers by coating ammonia-plasma treated HDPE with NCO-sP(EO-stat-PO), functionalization with linear RGD-peptides, and augmentation with RGD and BMP-4 was analyzed. Therefore, implants were placed subcutaneously in the paravertebral tissue and transcortically in the distal femur of another 14 Wistar rats. Both tests revealed no signs of enhanced inflammation of the surrounding tissue analyzed by CD68, IL-1ß-/TNF-α-antibody staining, nor systemic toxic reactions according to histological analysis of various organs. The mean thickness of the fibrous tissue surrounding the femoral implants was highest in native HDPE-implants and tended to be lower in all NCO-sP(EO-stat-PO) modified implants. Micro-CT analysis revealed a significant increase of peri-implant bone volume in RGD/BMP-4 coated samples. These results demonstrate that even very low amounts of surface bound growth factors do have significant effects when immobilized in an environment that retains their biological function. Hence, NCO-sP(EO-stat-PO)-coatings could offer an attractive platform to improve integration of orthopedic implants.


Assuntos
Materiais Revestidos Biocompatíveis/química , Hidrogéis/química , Oligopeptídeos/química , Polietilenoglicóis/química , Adsorção , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Proteína Morfogenética Óssea 4/química , Adesão Celular , Fêmur/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-1beta/metabolismo , Osseointegração , Próteses e Implantes , Ratos , Ratos Wistar , Propriedades de Superfície , Fatores de Tempo , Microtomografia por Raio-X
5.
J Trauma Acute Care Surg ; 74(2): 531-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23354247

RESUMO

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


Assuntos
Calo Ósseo/fisiopatologia , Fraturas do Fêmur/fisiopatologia , Síndrome de Resposta Inflamatória Sistêmica/etiologia , Traumatismos Torácicos/complicações , Animais , Calo Ósseo/química , Calo Ósseo/citologia , Calo Ósseo/patologia , Modelos Animais de Doenças , Interleucina-6/análise , Interleucina-6/fisiologia , Macrófagos/fisiologia , Masculino , Traumatismo Múltiplo/fisiopatologia , Neutrófilos/fisiologia , Osteoclastos/fisiologia , Ratos , Ratos Wistar , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Fatores de Tempo
6.
Immunobiology ; 218(1): 1-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22464814

RESUMO

The skeletal and the immune system are not two independent systems, rather, there are multifaceted and complex interactions between the different cell types of both systems and there are several shared cytokines. As a part of the innate immunity, the complement system was found to be an important link between bone and immunity. Complement proteins appear to be involved in bone development and homeostasis, and specifically influence osteoblast and osteoclast activity. This review describes the complex mutual regulation of the two systems, and indicates some of the negative side effects as a result of inappropriate or excessive complement activation.


Assuntos
Desenvolvimento Ósseo/imunologia , Regeneração Óssea/imunologia , Proteínas do Sistema Complemento/fisiologia , Osteoblastos/imunologia , Osteoclastos/imunologia , Animais , Ativação do Complemento , Citocinas/fisiologia , Humanos , Sistema Imunitário/fisiologia , Imunidade Inata , Sistema Musculoesquelético/imunologia
7.
J Orthop Res ; 31(3): 465-71, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23070742

RESUMO

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


Assuntos
Pinos Ortopédicos , Conversão para Cirurgia Aberta/efeitos adversos , Fixadores Externos , Fraturas do Fêmur/cirurgia , Consolidação da Fratura/fisiologia , Animais , Fenômenos Biomecânicos/fisiologia , Complemento C5a/metabolismo , Modelos Animais de Doenças , Fraturas do Fêmur/diagnóstico por imagem , Fraturas do Fêmur/fisiopatologia , Masculino , Osteotomia/métodos , Ratos , Ratos Wistar , Índices de Gravidade do Trauma , Ferimentos não Penetrantes/diagnóstico por imagem , Ferimentos não Penetrantes/fisiopatologia , Ferimentos não Penetrantes/cirurgia , Microtomografia por Raio-X
8.
Nat Rev Rheumatol ; 8(3): 133-43, 2012 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22293759

RESUMO

Optimal fracture treatment requires knowledge of the complex physiological process of bone healing. The course of bone healing is mainly influenced by fracture fixation stability (biomechanics) and the blood supply to the healing site (revascularization after trauma). The repair process proceeds via a characteristic sequence of events, described as the inflammatory, repair and remodeling phases. An inflammatory reaction involving immune cells and molecular factors is activated immediately in response to tissue damage and is thought to initiate the repair cascade. Immune cells also have a major role in the repair phase, exhibiting important crosstalk with bone cells. After bony bridging of the fragments, a slow remodeling process eventually leads to the reconstitution of the original bone structure. Systemic inflammation, as observed in patients with rheumatoid arthritis, diabetes mellitus, multiple trauma or sepsis, can increase fracture healing time and the rate of complications, including non-unions. In addition, evidence suggests that insufficient biomechanical conditions within the fracture zone can influence early local inflammation and impair bone healing. In this Review, we discuss the main factors that influence fracture healing, with particular emphasis on the role of inflammation.


Assuntos
Consolidação da Fratura/fisiologia , Inflamação/fisiopatologia , Animais , Fenômenos Biomecânicos , Remodelação Óssea/fisiologia , Modelos Animais de Doenças , Fixação de Fratura/métodos , Humanos , Ovinos
9.
J Orthop Res ; 30(4): 581-6, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21922535

RESUMO

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


Assuntos
Fraturas do Fêmur/imunologia , Consolidação da Fratura/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Traumatismos Torácicos/imunologia , Ferimentos não Penetrantes/imunologia , Animais , Fenômenos Biomecânicos/fisiologia , Complemento C5a/antagonistas & inibidores , Complemento C5a/imunologia , Modelos Animais de Doenças , Fraturas do Fêmur/diagnóstico por imagem , Fraturas do Fêmur/fisiopatologia , Consolidação da Fratura/imunologia , Inflamação/complicações , Inflamação/tratamento farmacológico , Inflamação/fisiopatologia , Masculino , Osteotomia , Ratos , Ratos Wistar , Receptor da Anafilatoxina C5a/imunologia , Traumatismos Torácicos/fisiopatologia , Índices de Gravidade do Trauma , Ferimentos não Penetrantes/fisiopatologia , Microtomografia por Raio-X
10.
J Cell Biochem ; 112(9): 2594-605, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21598302

RESUMO

There is a tight interaction of the bone and the immune system. However, little is known about the relevance of the complement system, an important part of innate immunity and a crucial trigger for inflammation. The aim of this study was, therefore, to investigate the presence and function of complement in bone cells including osteoblasts, mesenchymal stem cells (MSC), and osteoclasts. qRT-PCR and immunostaining revealed that the central complement receptors C3aR and C5aR, complement C3 and C5, and membrane-bound regulatory proteins CD46, CD55, and CD59 were expressed in human MSC, osteoblasts, and osteoclasts. Furthermore, osteoblasts and particularly osteoclasts were able to activate complement by cleaving C5 to its active form C5a as measured by ELISA. Both C3a and C5a alone were unable to trigger the release of inflammatory cytokines interleukin (IL)-6 and IL-8 from osteoblasts. However, co-stimulation with the pro-inflammatory cytokine IL-1ß significantly induced IL-6 and IL-8 expression as well as the expression of receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin (OPG) indicating that complement may modulate the inflammatory response of osteoblastic cells in a pro-inflammatory environment as well as osteoblast-osteoclast interaction. While C3a and C5a did not affect osteogenic differentiation, osteoclastogenesis was significantly induced even in the absence of RANKL and macrophage-colony stimulating factor (M-CSF) suggesting that complement could directly regulate osteoclast formation. It can therefore be proposed that complement may enhance the inflammatory response of osteoblasts and increase osteoclast formation, particularly in a pro-inflammatory environment, for example, during bone healing or in inflammatory bone disorders.


Assuntos
Complemento C3a/farmacologia , Complemento C5a/farmacologia , Interleucina-1beta/farmacologia , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Adulto , Antígenos CD/genética , Antígenos CD/metabolismo , Diferenciação Celular , Células Cultivadas , Complemento C3a/metabolismo , Complemento C3a/fisiologia , Complemento C5a/metabolismo , Complemento C5a/fisiologia , Expressão Gênica , Humanos , Inflamação , Interleucina-1beta/fisiologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Microscopia de Fluorescência , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese , Osteoprotegerina/metabolismo , Proteólise , Ligante RANK/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Adulto Jovem
11.
Acta Orthop ; 82(2): 223-7, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21463222

RESUMO

BACKGROUND AND PURPOSE: There is some clinical evidence that fracture healing is impaired in multiply injured patients. Nothing is known, however, about the effects of various types of injuries and their contribution to a possible disturbance of the fracture-healing process. We investigated the effect of a thoracic trauma and an additional soft-tissue trauma on fracture healing in a rat tibia model. METHODS: 3 groups of rats were operated: group A with a simple fracture of the tibia and fibula, group B with a fracture and an additional thoracic trauma, and group C with a fracture, thoracic trauma, and an additional soft-tissue trauma. The fracture and the soft-tissue injury were produced by a special guillotine-like device and the thoracic trauma by a blast wave generator. After one day, the serum level of IL-6 was quantified, and at the end of the study (28 days) the mechanical properties and the callus volume of the healed tibia were determined. RESULTS: Increasing the severity of the injury caused IL-6 levels to more than double 1 day after injury. It halved the load to failure in mechanical tests and led to reduced callus volume after 28 days of healing. INTERPRETATION: Fracture healing is impaired when additional thoracic trauma and soft tissue trauma occurs.


Assuntos
Consolidação da Fratura/fisiologia , Traumatismo Múltiplo/fisiopatologia , Lesões dos Tecidos Moles/fisiopatologia , Traumatismos Torácicos/fisiopatologia , Animais , Fíbula/lesões , Escala de Gravidade do Ferimento , Interleucina-6/sangue , Masculino , Modelos Biológicos , Ratos , Ratos Wistar , Lesões dos Tecidos Moles/complicações , Traumatismos Torácicos/complicações , Fraturas da Tíbia/complicações , Fraturas da Tíbia/fisiopatologia
12.
J Trauma ; 71(4): 952-60, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21460748

RESUMO

BACKGROUND: There is evidence that complement components regulate cytokine production in osteoblastic cells, induce cell migration in mesenchymal stem cells, and play a regulatory role in normal enchondral bone formation. We proved the hypothesis that complement might be involved in bone healing after fracture. METHODS: We investigated the expression of the key anaphylatoxin receptor C5aR during fracture healing in rats by immunostaining after 1, 3, 7, 14, and 28 days. C5aR expression was additionally analyzed in human mesenchymal stem cells (hMSC) during osteogenic differentiation, in human primary osteoblasts, and osteoclasts by reverse transcriptase polymerase chain reaction and immunostaining. Receptor functionality was proven by the migratory response of cells to C5a in a Boyden chamber. RESULTS: C5aR was expressed in a distinct spatial and temporal pattern in the fracture callus by differentiated osteoblast, chondroblast-like cells in cartilaginous regions, and osteoclasts. In vitro C5aR was expressed by osteoblasts, osteoclasts, and hMSC undergoing osteogenic differentiation. C5aR was barely expressed by undifferentiated hMSC but was significantly induced after osteogenic differentiation. C5aR activation by C5a induced strong chemotactic activity in osteoblasts, and in hMSC, which had undergone osteogenic differentiation, being abolished by a specific C5aR antagonist. In hMSC, C5a induced less migration reflecting their low level of C5aR expression. CONCLUSIONS: Our in vitro and in vivo results demonstrated the presence of C5aR in bone forming osteoblasts and bone resorbing osteoclasts. It is suggested that C5aR might play a regulatory role in fracture healing in intramembranous and in enchondral ossification, one possible function being the regulation of cell recruitment.


Assuntos
Movimento Celular/fisiologia , Consolidação da Fratura/fisiologia , Osteoblastos/fisiologia , Receptor da Anafilatoxina C5a/fisiologia , Adulto , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Masculino , Células-Tronco Mesenquimais/fisiologia , Osteoclastos/fisiologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Adulto Jovem
13.
J Orthop Res ; 29(5): 734-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21437953

RESUMO

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


Assuntos
Fraturas do Fêmur/terapia , Consolidação da Fratura/fisiologia , Traumatismos Torácicos/fisiopatologia , Ferimentos não Penetrantes/fisiopatologia , Animais , Fenômenos Biomecânicos , Fixadores Externos , Fraturas do Fêmur/cirurgia , Inflamação/etiologia , Interleucina-6/sangue , Masculino , Ratos , Ratos Wistar , Traumatismos Torácicos/complicações , Ferimentos não Penetrantes/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...