Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Trauma Case Rep ; 42: 100718, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36281425

RESUMEN

Nowadays, massive segmental bone defects represent a surgical challenge for trauma surgeons. Most of these injuries appear in the context of high-energy trauma, not only significantly affecting the bones, but also involving severe injuries of the adjacent soft tissues. For these reasons, their treatment requires complex reconstruction surgeries. There are multiple techniques to treat bone defects, bone transport being one of the most widely used. Historically, external fixators (monolateral and circular) have been and still are the gold standard for performing this technique, although they are not exempt from complications. By means of specific intramedullary nails for bone transport, it is possible to minimize the complications of external fixation, allowing large tibial bone defects to be treated through distraction osteogenesis (all-internal system), which is favoured by early weight bearing.

2.
J Tissue Eng Regen Med ; 13(5): 742-752, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30785671

RESUMEN

An attractive alternative to bone autografts is the use of autologous mesenchymal progenitor cells (MSCs) in combination with biomaterials. We compared the therapeutic potential of different sources of mesenchymal stem cells in combination with biomaterials in a bone nonunion model. A critical-size defect was created in Sprague-Dawley rats. Animals were divided into six groups, depending on the treatment to be applied: bone defect was left empty (CTL); treated with live bone allograft (LBA); hrBMP-2 in collagen scaffold (CSBMP2 ); acellular polycaprolactone scaffold (PCL group); PCL scaffold containing periosteum-derived MSCs (PCLPMSCs ) and PCL containing bone marrow-derived MSCs (PCLBMSCs ). To facilitate cell tracking, both MSCs and bone graft were isolated from green fluorescent protein (GFP)-transgenic rats. CTL group did not show any signs of healing during the radiological follow-up (n = 6). In the LBA group, all the animals showed bone bridging (n = 6) whereas in the CSBMP2 group, four out of six animals demonstrated healing. In PCL and PCLPMSCs groups, a reduced number of animals showed radiological healing, whereas no healing was detected in the PCLBMSCs group. Using microcomputed tomography, the bone volume filling the defect was quantified, showing significant new bone formation in the LBA, CSBMP2 , and PCLPMSCs groups when compared with the CTL group. At 10 weeks, GFP positive cells were detected only in the LBA group and restricted to the outer cortical bone in close contact with the periosteum. Tracking of cellular implants demonstrated significant survival of the PMSCs when compared with BMSCs. In conclusion, PMSCs improve bone regeneration being suitable for mimetic autograft design.


Asunto(s)
Bioprótesis , Fracturas del Fémur/terapia , Curación de Fractura , Células Madre Mesenquimatosas/metabolismo , Periostio/metabolismo , Ingeniería de Tejidos , Animales , Fracturas del Fémur/metabolismo , Fracturas del Fémur/patología , Células Madre Mesenquimatosas/patología , Periostio/patología , Ratas , Ratas Sprague-Dawley
3.
Stem Cells ; 34(9): 2342-53, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27250101

RESUMEN

Fracture nonunion is a major complication of bone fracture regeneration and repair. The molecular mechanisms that result in fracture nonunion appearance are not fully determined. We hypothesized that fracture nonunion results from the failure of hypoxia and hematoma, the primary signals in response to bone injury, to trigger Bmp2 expression by mesenchymal progenitor cells (MSCs). Using a model of nonstabilized fracture healing in transgenic 5'Bmp2BAC mice we determined that Bmp2 expression appears in close association with hypoxic tissue and hematoma during the early phases of fracture healing. In addition, BMP2 expression is induced when human periosteum explants are exposed to hypoxia ex vivo. Transient interference of hypoxia signaling in vivo with PX-12, a thioredoxin inhibitor, results in reduced Bmp2 expression, impaired fracture callus formation and atrophic-like nonunion by a HIF-1α independent mechanism. In isolated human periosteum-derived MSCs, BMP2 expression could be induced with the addition of platelets concentrate lysate but not with hypoxia treatment, confirming HIF-1α-independent BMP2 expression. Interestingly, in isolated human periosteum-derived mesenchymal progenitor cells, inhibition of BMP2 expression by PX-12 is accomplished only under hypoxic conditions seemingly through dis-regulation of reactive oxygen species (ROS) levels. In conclusion, we provide evidence of a molecular mechanism of hypoxia-dependent BMP2 expression in MSCs where interference with ROS homeostasis specifies fracture nonunion-like appearance in vivo through inhibition of Bmp2 expression. Stem Cells 2016;34:2342-2353.


Asunto(s)
Fracturas no Consolidadas/metabolismo , Fracturas no Consolidadas/patología , Homeostasis , Células Madre Mesenquimatosas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Proteína Morfogenética Ósea 2/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Separación Celular , Disulfuros/farmacología , Curación de Fractura/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Imidazoles/farmacología , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Osteogénesis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Periostio/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA