Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cells Int ; 2016: 6530624, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26770211

RESUMEN

Osteoporotic patients, incapacitated due to vertebral compression fractures (VCF), suffer grave financial and clinical burden. Current clinical treatments focus on symptoms' management but do not combat the issue at the source. In this pilot study, allogeneic, porcine mesenchymal stem cells, overexpressing the BMP6 gene (MSC-BMP6), were suspended in fibrin gel and implanted into a vertebral defect to investigate their effect on bone regeneration in a clinically relevant, large animal pig model. To check the effect of the BMP6-modified cells on bone regeneration, a fibrin gel only construct was used for comparison. Bone healing was evaluated in vivo at 6 and 12 weeks and ex vivo at 6 months. In vivo CT showed bone regeneration within 6 weeks of implantation in the MSC-BMP6 group while only minor bone formation was seen in the defect site of the control group. After 6 months, ex vivo analysis demonstrated enhanced bone regeneration in the BMP6-MSC group, as compared to control. This preclinical study presents an innovative, potentially minimally invasive, technique that can be used to induce bone regeneration using allogeneic gene modified MSCs and therefore revolutionize current treatment of challenging conditions, such as osteoporosis-related VCFs.

2.
J Vis Exp ; (106): e53459, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26779586

RESUMEN

A major parameter determining the success of a bone-grafting procedure is vascularization of the area surrounding the graft. We hypothesized that implantation of a bone autograft would induce greater bone regeneration by abundant blood vessel formation. To investigate the effect of the graft on neovascularization at the defect site, we developed a micro-computed tomography (µCT) approach to characterize newly forming blood vessels, which involves systemic perfusion of the animal with a polymerizing contrast agent. This method enables detailed vascular analysis of an organ in its entirety. Additionally, blood perfusion was assessed using fluorescence imaging (FLI) of a blood-borne fluorescent agent. Bone formation was quantified by FLI using a hydroxyapatite-targeted probe and µCT analysis. Stem cell recruitment was monitored by bioluminescence imaging (BLI) of transgenic mice that express luciferase under the control of the osteocalcin promoter. Here we describe and demonstrate preparation of the allograft, calvarial defect surgery, µCT scanning protocols for the neovascularization study and bone formation analysis (including the in vivo perfusion of contrast agent), and the protocol for data analysis. The 3D high-resolution analysis of vasculature demonstrated significantly greater angiogenesis in animals with implanted autografts, especially with respect to arteriole formation. Accordingly, blood perfusion was significantly higher in the autograft group by the 7(th) day after surgery. We observed superior bone mineralization and measured greater bone formation in animals that received autografts. Autograft implantation induced resident stem cell recruitment to the graft-host bone suture, where the cells differentiated into bone-forming cells between the 7(th) and 10(th) postoperative day. This finding means that enhanced bone formation may be attributed to the augmented vascular feeding that characterizes autograft implantation. The methods depicted may serve as an optimal tool to study bone regeneration in terms of tightly bounded bone formation and neovascularization.


Asunto(s)
Aloinjertos/anatomía & histología , Regeneración Ósea/fisiología , Trasplante Óseo/métodos , Huesos/irrigación sanguínea , Cráneo/trasplante , Aloinjertos/irrigación sanguínea , Aloinjertos/diagnóstico por imagen , Animales , Autoinjertos/irrigación sanguínea , Autoinjertos/diagnóstico por imagen , Huesos/anatomía & histología , Huesos/diagnóstico por imagen , Diferenciación Celular , Femenino , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/fisiología , Imagen Óptica/métodos , Osteogénesis/efectos de los fármacos , Cráneo/anatomía & histología , Cráneo/irrigación sanguínea , Cráneo/diagnóstico por imagen , Microtomografía por Rayos X/métodos
3.
Mol Pharm ; 10(12): 4462-71, 2013 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-24131143

RESUMEN

Allografts may be useful in craniofacial bone repair, although they often fail to integrate with the host bone. We hypothesized that intermittent administration of parathyroid hormone (PTH) would enhance mesenchymal stem cell recruitment and differentiation, resulting in allograft osseointegration in cranial membranous bones. Calvarial bone defects were created in transgenic mice, in which luciferase is expressed under the control of the osteocalcin promoter. The mice were given implants of allografts with or without daily PTH treatment. Bioluminescence imaging (BLI) was performed to monitor host osteprogenitor differentiation at the implantation site. Bone formation was evaluated with the aid of fluorescence imaging (FLI) and microcomputed tomography (µCT) as well as histological analyses. Reverse transcription polymerase chain reaction (RT-PCR) was performed to evaluate the expression of key osteogenic and angiogenic genes. Osteoprogenitor differentiation, as detected by BLI, in mice treated with an allograft implant and PTH was over 2-fold higher than those in mice treated with an allograft implant without PTH. FLI also demonstrated that the bone mineralization process in PTH-treated allografts was significantly higher than that in untreated allografts. The µCT scans revealed a significant increase in bone formation in allograft + PTH treated mice comparing to allograft + PBS treated mice. The osteogenic genes osteocalcin (Oc/Bglap) and integrin binding sialoprotein (Ibsp) were upregulated in the allograft + PTH treated animals. In summary, PTH treatment enhances osteoprogenitor differentiation and augments bone formation around structural allografts. The precise mechanism is not clear, but we show that infiltration pattern of mast cells, associated with the formation of fibrotic tissue, in the defect site is significantly affected by the PTH treatment.


Asunto(s)
Huesos/efectos de los fármacos , Huesos/fisiología , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Hormona Paratiroidea/farmacología , Aloinjertos/efectos de los fármacos , Aloinjertos/fisiología , Animales , Trasplante Óseo/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Expresión Génica/fisiología , Mastocitos/efectos de los fármacos , Mastocitos/fisiología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Osteocalcina/genética , Osteogénesis/genética , Regiones Promotoras Genéticas/genética , Sialoglicoproteínas/genética , Trasplante Homólogo/métodos
4.
Methods Mol Biol ; 1048: 181-93, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23929106

RESUMEN

Real-time bioluminescence functional imaging holds great promise for regenerative medicine because it improves the researcher's ability to analyze and understand the healing process. Using transgenic mice coupled with gene-modified cells, one can employ this method to monitor host and graft activity in various models of tissue regeneration. We implemented real-time bioluminescence functional imaging to analyze bone formation by following a unique protocol in which the luciferase reporter gene, driven by an osteocalcin promoter, is used to visualize host and graft activity during bone formation. Real-time bioluminescence functional imaging can be used to assess the "host reaction" in transgenic mice models; it can also be used to assess "graft activity" in other animals in which genetically labeled stem cells have been implanted or direct gene delivery has been applied. The suggested imaging protocol requires 25 min per sample. However, special attention must be given to the layout of the experimental design, which determines the specific activity that will be analyzed.


Asunto(s)
Regeneración Ósea , Regeneración Tisular Dirigida/métodos , Mediciones Luminiscentes , Ingeniería de Tejidos/métodos , Animales , Técnicas de Transferencia de Gen , Luciferasas/genética , Ratones , Ratones Transgénicos , Osteocalcina/genética , Osteogénesis/fisiología , Regiones Promotoras Genéticas/genética
5.
Tissue Eng Part A ; 19(5-6): 748-58, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23215901

RESUMEN

Osteogenesis of mesenchymal stem cells (MSCs) is highly dependent on oxygen supply. We have shown that perfluorotributylamine (PFTBA), a synthetic oxygen carrier, enhances MSC-based bone formation in vivo. Exploring this phenomenon's mechanism, we hypothesize that a transient increase in oxygen levels using PFTBA will affect MSC survival, proliferation, and differentiation, thus increasing bone formation. To test this hypothesis, MSCs overexpressing bone morphogenetic protein 2 were encapsulated in alginate beads that had been supplemented with an emulsion of PFTBA or phosphate-buffered saline. Oxygen measurements showed that supplementation of PFTBA significantly increased the available oxygen level during a 96-h period. PFTBA-containing beads displayed an elevation in cell viability, which was preserved throughout 2 weeks, and a significantly lower ratio of dead cells throughout the experiment. Furthermore, the cells from the control group expressed significantly more hypoxia-related genes such as VEGF, DDIT3, and PKG1. Additionally, PFTBA supplementation led to an increase in the osteogenic differentiation and to a decrease in chondrogenic differentiation of MSCs. In conclusion, PFTBA increases the oxygen availability in the vicinity of the MSCs, which suffer oxygen exhaustion shortly after encapsulation in alginate beads. Consequently, cell survival is increased, and hypoxia-related genes are downregulated. In addition, PFTBA promotes osteogenic differentiation over chondrogeneic differentiation, and thereby can accelerate MSC-based bone regeneration.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/citología , Osteogénesis , Oxígeno/farmacología , Animales , Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Condrogénesis/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Femenino , Fluorocarburos/farmacología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteogénesis/efectos de los fármacos , Tejido Subcutáneo/efectos de los fármacos
6.
Adv Drug Deliv Rev ; 64(12): 1320-30, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22429662

RESUMEN

Bone formation and regeneration therapies continue to require optimization and improvement because many skeletal disorders remain undertreated. Clinical solutions to nonunion fractures and osteoporotic vertebral compression fractures, for example, remain suboptimal and better therapeutic approaches must be created. The widespread use of recombinant human bone morphogenetic proteins (rhBMPs) for spine fusion was recently questioned by a series of reports in a special issue of The Spine Journal, which elucidated the side effects and complications of direct rhBMP treatments. Gene therapy - both direct (in vivo) and cell-mediated (ex vivo) - has long been studied extensively to provide much needed improvements in bone regeneration. In this article, we review recent advances in gene therapy research whose aims are in vivo or ex vivo bone regeneration or formation. We examine appropriate vectors, safety issues, and rates of bone formation. The use of animal models and their relevance for translation of research results to the clinical setting are also discussed in order to provide the reader with a critical view. Finally, we elucidate the main challenges and hurdles faced by gene therapy aimed at bone regeneration as well as expected future trends in this field.


Asunto(s)
Enfermedades Óseas/terapia , Regeneración Ósea/genética , Terapia Genética/métodos , Animales , Enfermedades Óseas/patología , Proteínas Morfogenéticas Óseas/administración & dosificación , Proteínas Morfogenéticas Óseas/efectos adversos , Proteínas Morfogenéticas Óseas/uso terapéutico , Huesos/metabolismo , Huesos/patología , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Humanos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Fusión Vertebral/métodos , Ingeniería de Tejidos/métodos
7.
J Struct Biol ; 177(2): 314-28, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22248452

RESUMEN

This study investigates the three-dimensional structure of the eight plate exoskeletal (shell) assembly of the chiton Tonicella marmorea. X-ray micro-computed tomography and 3D printing elucidate the mechanism of conformational change from a passive (slightly curved, attached to surface) to a defensive (rolled, detached from surface) state of the plate assembly. The passive and defensive conformations exhibited differences in longitudinal curvature index (0.43 vs. 0.70), average plate-to-plate overlap (∼62% vs. ∼48%), cross-sectional overlap heterogeneity (60-82.5% vs. 0-90%, fourth plate), and plate-to-plate separation distance (100% increase in normalized separation distance between plates 4 and 5), respectively. The plate-to-plate interconnections consist of two rigid plates joined by a compliant, actuating muscle, analogous to a geometrically structured shear lap joint. This work provides an understanding of how T. marmorea achieves the balance between mobility and protection. In the passive state, the morphometry of the plates and plate-to-plate interconnections results in an approximately continuous curvature and constant armor thickness, resulting in limited mobility but maximum protection. In the defensive state, the underlying soft tissues gain protection and the chiton gains mobility through tidal flow, but regions of vulnerability open dorsally, due to the increase in plate-to-plate separation and decrease in plate-to-plate overlap. Lastly, experiments using optical and scanning electron microscopy, mercury porosimetry, and Fourier-transform infrared spectroscopy explore the microstructure and spatial distribution of the six layers within the intermediate plates, the role of multilayering in resisting predatory attacks, and the detection of chitin as a major component of the intra-plate organic matrix and girdle.


Asunto(s)
Exoesqueleto/anatomía & histología , Poliplacóforos/anatomía & histología , Exoesqueleto/metabolismo , Animales , Fenómenos Biomecánicos , Carbonato de Calcio/metabolismo , Modelos Anatómicos , Modelos Biológicos , Poliplacóforos/metabolismo , Microtomografía por Rayos X
8.
Mol Pharm ; 8(5): 1592-601, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21834548

RESUMEN

Vertebral compression fractures (VCFs), the most common fragility fractures, account for approximately 700,000 injuries per year. Since open surgery involves morbidity and implant failure in the osteoporotic patient population, a new minimally invasive biological solution to vertebral bone repair is needed. Previously, we showed that adipose-derived stem cells (ASCs) overexpressing a BMP gene are capable of inducing spinal fusion in vivo. We hypothesized that a direct injection of ASCs, designed to transiently overexpress rhBMP6, into a vertebral bone void defect would accelerate bone regeneration. Porcine ASCs were isolated and labeled with lentiviral vectors that encode for the reporter gene luciferase (Luc) under constitutive (ubiquitin) or inductive (osteocalcin) promoters. The ASCs were first labeled with reporter genes and then nucleofected with an rhBMP6-encoding plasmid. Twenty-four hours later, bone void defects were created in the coccygeal vertebrae of nude rats. The ASC-BMP6 cells were suspended in fibrin gel (FG) and injected into the bone void. A control group was injected with FG alone. The regenerative process was monitored in vivo using microCT, and cell survival and differentiation were monitored using tissue specific reporter genes and bioluminescence imaging (BLI). The surgically treated vertebrae were harvested after 12 weeks and subjected to histological and immunohistochemical (against porcine vimentin) analyses. In vivo BLI detected Luc-expressing cells at the implantation site over a 12-week period. Beginning 2 weeks postoperatively, considerable defect repair was observed in the group treated with ASC-BMP6 cells. The rate of bone formation in the stem cell-treated group was two times faster than that in the FG-treated group, and bone volume at the end point was 2-fold compared to the control group. Twelve weeks after cell injection the bone volume within the void reached the volume measured in native vertebrae. Immunostaining against porcine vimentin indicated that the ASC-BMP6 cells contributed to new bone formation. Here we show the potential of injections of BMP-modified ASCs to repair vertebral bone defects in a rat model. Our results could pave the way to a novel approach for the biological treatment of traumatic and osteoporosis-related vertebral bone injuries.


Asunto(s)
Células Madre Adultas/trasplante , Proteína Morfogenética Ósea 6/uso terapéutico , Regeneración Ósea , Técnicas de Transferencia de Gen , Traumatismos Vertebrales/terapia , Columna Vertebral/fisiología , Células Madre Adultas/metabolismo , Animales , Proteína Morfogenética Ósea 6/genética , Proteína Morfogenética Ósea 6/metabolismo , Células Cultivadas , Fibrina/química , Genes Reporteros , Hidrogel de Polietilenoglicol-Dimetacrilato , Osteocalcina/genética , Regiones Promotoras Genéticas , Radiografía , Distribución Aleatoria , Ratas , Ratas Desnudas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Traumatismos Vertebrales/diagnóstico por imagen , Traumatismos Vertebrales/metabolismo , Traumatismos Vertebrales/patología , Columna Vertebral/diagnóstico por imagen , Columna Vertebral/patología , Grasa Subcutánea Abdominal/citología , Porcinos , Porcinos Enanos , Cola (estructura animal) , Ubiquitina/genética
9.
Nat Protoc ; 6(1): 105-10, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21212786

RESUMEN

Microcomputed tomography (microCT) analysis is a powerful tool for the evaluation of bone tissue because it provides access to the 3D microarchitecture of the bone. It is invaluable for regenerative medicine as it provides the researcher with the opportunity to explore the skeletal system both in vivo and ex vivo. The quantitative assessment of macrostructural characteristics and microstructural features may improve our ability to estimate the quality of newly formed bone. We have developed a unique procedure for analyzing data from microCT scans to evaluate bone structure and repair. This protocol describes the procedures for microCT analysis of three main types of mouse bone regeneration models (ectopic administration of bone-forming mesenchymal stem cells, and administration of cells after both long bone defects and cranial segmental bone defects) that can be easily adapted for a variety of other models. Precise protocols are crucial because the system is extremely user sensitive and results can be easily biased if standardized methods are not applied. The suggested protocol takes 1.5-3.5 h per sample, depending on bone tissue sample size, the type of equipment used, variables of the scanning protocol and the operator's experience.


Asunto(s)
Regeneración Ósea , Huesos/fisiología , Microtomografía por Rayos X/métodos , Animales , Huesos/patología , Huesos/ultraestructura , Ratones , Modelos Biológicos
10.
J Tissue Eng Regen Med ; 5(5): 384-93, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20740691

RESUMEN

Mechanical loading has been described as a highly important stimulus for improvements in the quality and strength of bone. It has also been shown that mechanical stimuli can induce the differentiation of mesenchymal stem cells (MSCs) along the osteogenic lineage. We have previously demonstrated the potent osteogenic effect of MSCs engineered to overexpress the BMP2 gene. In this study we investigated the effect of mechanical loading on BMP2-expressing MSC-like cells, using a special bioreactor designed to apply dynamic forces on cell-seeded hydrogels. Cell viability, alkaline phosphatase (ALP) activity, BMP2 secretion and mineralized substance formation in the hydrogels were quantified. We found that cell metabolism increased as high as 6.8-fold, ALP activity by 12.5-fold, BMP2 secretion by 182-fold and mineralized tissue formation by 1.72-fold in hydrogels containing MSC-like cells expressing BMP2, which were cultured in the presence of mechanical loading. We have shown that ex vivo mechanical loading had an additive effect on BMP2-induced osteogenesis in genetically engineered MSC-like cells. These data could be valuable for bone tissue-engineering strategies of the future.


Asunto(s)
Ingeniería Genética/métodos , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos/métodos , Fosfatasa Alcalina/metabolismo , Animales , Reactores Biológicos , Proteína Morfogenética Ósea 2/metabolismo , Huesos/metabolismo , Diferenciación Celular , Fibrinógeno/química , Hidrogeles , Ratones , Ratones Endogámicos C3H , Osteogénesis , Polietilenglicoles/química
11.
Mol Ther ; 19(1): 53-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20859259

RESUMEN

Nonunion fractures present a challenge to orthopedics with no optimal solution. In-vivo DNA electroporation is a gene-delivery technique that can potentially accelerate regenerative processes. We hypothesized that in vivo electroporation of an osteogenic gene in a nonunion radius bone defect site would induce fracture repair. Nonunion fracture was created in the radii of C3H/HeN mice, into which a collagen sponge was placed. To allow for recruitment of host progenitor cells (HPCs) into the implanted sponge, the mice were housed for 10 days before electroporation. Mice were electroporated with either bone morphogenetic protein 9 (BMP-9) plasmid, Luciferase plasmid or injected with BMP-9 plasmid but not electroporated. In vivo bioluminescent imaging indicated that gene expression was localized to the defect site. Microcomputed tomography (µCT) and histological analysis of murine radii electroporated with BMP-9 demonstrated bone formation bridging the bone gap, whereas in the control groups the defect remained unbridged. Population of the implanted collagen sponge by HPCs transfected with the injected plasmid following electroporation was noted. Our data indicate that regeneration of nonunion bone defect can be attained by performing in vivo electroporation with an osteogenic gene combined with recruitment of HPCs. This gene therapy approach may pave the way for regeneration of other skeletal tissues.


Asunto(s)
Regeneración Ósea/genética , Electroporación/métodos , Fracturas no Consolidadas/terapia , Terapia Genética/métodos , Factor 2 de Diferenciación de Crecimiento/genética , Osteogénesis/genética , Células Madre/fisiología , Animales , Colágeno/administración & dosificación , Femenino , Fracturas no Consolidadas/patología , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Factor 2 de Diferenciación de Crecimiento/biosíntesis , Luciferasas/biosíntesis , Luciferasas/genética , Ratones , Ratones Endogámicos C3H , Plásmidos/genética , Cicatrización de Heridas/genética
12.
Tissue Eng Part A ; 16(12): 3679-86, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20618082

RESUMEN

Most spine fusion procedures involve the use of prosthetic fixation devices combined with autologous bone grafts rather than biological treatment. We had shown that spine fusion could be achieved by injection of bone morphogenetic protein-2 (BMP-2)-expressing mesenchymal stem cells (MSCs) into the paraspinal muscle. In this study, we hypothesized that posterior spinal fusion achieved using genetically modified MSCs would be mechanically comparable to that realized using a mechanical fixation. BMP-2-expressing MSCs were injected bilaterally into paravertebral muscles of the mouse lumbar spine. In one control group BMP-2 expression was inhibited. Microcomputed tomography and histological analyses were used to evaluate bone formation. For comparison, a group of mouse spines were bilaterally fused with stainless steel pins. The harvested spines were later tested using a custom four-point bending apparatus and structural bending stiffness was estimated. To assess the degree to which MSC vertebral fusion was targeted and to quantify the effects of fusion on adjacent spinal segments, images of the loaded spine curvature were analyzed to extract rigidity of the individual spinal segments. Bone bridging of the targeted vertebrae was observed in the BMP-2-expressing MSC group, whereas no bone formation was noted in any control group. The biomechanical tests showed that MSC-mediated spinal fusion was as effective as stainless steel pin-based fusion and significantly more rigid than the control groups. Local analysis showed that the distribution of stiffness in the MSC-based fusion group was similar to that in the steel pin fusion group, with the majority of spinal stiffness contributed by the targeted fusion at L3-L5. Our findings demonstrate that MSC-induced spinal fusion can convey biomechanical rigidity to a targeted segment that is comparable to that achieved using an instrumental fixation.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Fusión Vertebral , Animales , Fenómenos Biomecánicos , Línea Celular , Femenino , Inmunohistoquímica , Células Madre Mesenquimatosas/fisiología , Ratones , Osteogénesis/genética , Osteogénesis/fisiología , Columna Vertebral/citología , Columna Vertebral/cirugía , Microtomografía por Rayos X
13.
J Biomech ; 43(12): 2315-20, 2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-20471652

RESUMEN

Stem cell-mediated gene therapy for fracture repair, utilizes genetically engineered mesenchymal stem cells (MSCs) for the induction of bone growth and is considered a promising approach in skeletal tissue regeneration. Previous studies have shown that murine nonunion fractures can be repaired by implanting MSCs over-expressing recombinant human bone morphogenetic protein-2 (rhBMP-2). Nanoindentation studies of bone tissue induced by MSCs in a radius fracture site indicated similar elastic modulus compared to intact murine bone, eight weeks post-treatment. In the present study we sought to investigate temporal changes in microarchitecture and biomechanical properties of repaired murine radius bones, following the implantation of MSCs. High-resolution micro-computed tomography (micro-CT) was performed 10 and 35 weeks post MSC implantation, followed by micro-finite element (micro-FE) analysis. The results have shown that the regenerated bone tissue remodels over time, as indicated by a significant decrease in bone volume, total volume, and connectivity density combined with an increase in mineral density. In addition, the axial stiffness of limbs repaired with MSCs was 2-1.5 times higher compared to the contralateral intact limbs, at 10 and 35 weeks post-treatment. These results could be attributed to the fusion that occurred in between the ulna and radius bones. In conclusion, although MSCs induce bone formation, which exceeds the fracture site, significant remodeling of the repair callus occurs over time. In addition, limbs treated with an MSC graft demonstrated superior biomechanical properties, which could indicate the clinical benefit of future MSC application in nonunion fracture repair.


Asunto(s)
Fracturas no Consolidadas/terapia , Terapia Genética/métodos , Trasplante de Células Madre Mesenquimatosas , Animales , Fenómenos Biomecánicos , Densidad Ósea , Proteína Morfogenética Ósea 2/genética , Regeneración Ósea , Remodelación Ósea , Modelos Animales de Enfermedad , Módulo de Elasticidad , Femenino , Análisis de Elementos Finitos , Fracturas no Consolidadas/diagnóstico por imagen , Fracturas no Consolidadas/fisiopatología , Ingeniería Genética , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C3H , Fracturas del Radio/diagnóstico por imagen , Fracturas del Radio/fisiopatología , Fracturas del Radio/terapia , Proteínas Recombinantes/genética , Microtomografía por Rayos X
14.
J Struct Biol ; 171(3): 318-31, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20433929

RESUMEN

In this study, a quantitative investigation of the microstructure and composition of field-caught marine Gasterosteus aculeatus (threespine stickleback) armor is presented, which provides useful phylogenetic information and insights into biomechanical function. Micro-computed tomography (microCT) was employed to create full three-dimensional images of the dorsal spines and basal plate, lateral plates, pelvic girdle and spines and to assess structural and compositional properties such as the spatial distribution of thickness (approximately 100-300 microm), the heterogeneous cross-sectional geometry (centrally thickened), plate-to-plate juncture and overlap (approximately 50% of the plate width), and bone mineral density (634-748 HA/cm(3)). The convolution of plate geometry in conjunction with plate-to-plate overlap allows a relatively constant armor thickness to be maintained throughout the assembly, promoting spatially homogeneous protection and thereby avoiding weakness at the armor unit interconnections. Plate-to-plate junctures act to register and join the plates while permitting compliance in sliding and rotation in selected directions. Mercury porosimetry was used to determine the pore size distribution and volume percent porosity of the lateral plates (20-35 vol.%) and spines (10-15 vol.%). SEM and microCT revealed a porous, sandwich-like cross-section beneficial for bending stiffness and strength at minimum weight. Back-scattered electron microscopy and energy dispersive X-ray analysis were utilized to quantify the weight percent mineral content (58-68%). Scanning electron microscopy and surface profilometry were used to characterize the interior and exterior surface topography (tubercles) of the lateral plates. The results obtained in this study are discussed in the context of mechanical function, performance, fitness, and survivability.


Asunto(s)
Estructuras Animales/anatomía & histología , Smegmamorpha/anatomía & histología , Estructuras Animales/química , Estructuras Animales/ultraestructura , Animales , Microscopía Electrónica de Rastreo , Espectrometría por Rayos X , Espectroscopía Infrarroja por Transformada de Fourier , Difracción de Rayos X , Microtomografía por Rayos X
15.
Bone ; 45(3): 414-22, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19481620

RESUMEN

Osteoarthritis (OA) affects both cartilage and bone tissues, and the subsequent breakdown of the two tissues appears to be interrelated. The interest in the role of subchondral bone changes with OA is growing, and one suggestion is that a simple inverse correlation exists between the cartilage loss and increased bone mineral density. In this work the STR/ort mouse is used as a model for human OA, in order to investigate disease progression. The aim of the work is to elucidate the tempero-spatial relationships between bone and cartilage architecture and determine whether a simple inverse correlation is satisfactory. We employ 3D whole joint quantitative imaging techniques for assessment of subchondral bone and articular cartilage. The knee joints of mice aged 3, 4, 7 and 10 months are scanned with muCT and then the tibial plateaus are scanned with CLSM. The results show that depending on site (medial and lateral), compartment (epiphyseal, metaphyseal, cortical), and age (3, 4, 7, 10 months), the subchondral bone undergoes changes that lead to an altered architecture. This is primarily seen as densification of the cortex and epiphysis in the STR/ort mice, with a significant change occurring between 7 and 10 months, while the medial cartilage thickness is significantly reduced after 7 months. Using a novel multimodal imaging approach, morphometric changes in the murine osteoarthritic knee joint are elucidated. It is seen that a complex interplay of events - both spatially and temporally - is involved in OA onset and progression. The initial measured differences between the two strains suggest a possible morphological phenotype involved in OA resistance/vulnerability. Temporally the changes have a strong strain:age dependence, although no separate timeline of events between the two tissues could be discerned. Spatially, the changes to medial and lateral morphometry across the cartilage and bone, indicate a relationship to altered joint mechanics.


Asunto(s)
Huesos/patología , Cartílago Articular/patología , Osteoartritis/patología , Envejecimiento/patología , Animales , Huesos/diagnóstico por imagen , Cartílago Articular/diagnóstico por imagen , Imagenología Tridimensional , Articulación de la Rodilla/patología , Masculino , Ratones , Microscopía Confocal , Osteoartritis/diagnóstico por imagen , Microtomografía por Rayos X
16.
Biomaterials ; 30(27): 4639-48, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19540585

RESUMEN

A major hurdle to surmount in bone-tissue engineering is ensuring a sufficient oxygen supply to newly forming tissue to avoid cell death or delayed development of osteogenic features. We hypothesized that an oxygen-enriched hydrogel scaffold would enhance tissue-engineered bone formation in vivo. To test this, we used a well-characterized mesenchymal stem cell (MSC) line, Tet-off BMP2 MSC, whose cells were engineered to express recombinant human bone morphogenetic protein-2. Cells were suspended in hydrogel supplemented with perfluorotributylamine (PFTBA) and implanted subcutaneously in an ectopic site, a radial bone defect, or a lumbar paravertebral muscle (mouse model of spinal fusion) in C3H/HeN mice. For controls, we used cells suspended in the same gel without PFTBA. In the ectopic site, there were significant increases in bone formation (2.5-fold increase), cell survival, and osteocalcin activity in the PFTBA-supplemented groups. PFTBA supplementation significantly increased structural parameters of bone in radial bone defects and triggered a significant 1.4-fold increase in bone volume in the spinal fusion model. We conclude that synthetic oxygen carrier supplementation of tissue-engineered implants enhances ectopic bone formation and yields better bone quality and volume in bone-repair and spinal fusion models, probably due to increased cell survival.


Asunto(s)
Fluorocarburos/farmacología , Hidrogeles/química , Células Madre Mesenquimatosas/citología , Osteogénesis/efectos de los fármacos , Oxígeno/metabolismo , Animales , Proteína Morfogenética Ósea 2/metabolismo , Regeneración Ósea/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Implantes Experimentales , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Osteocalcina/genética , Osteocalcina/metabolismo , Comunicación Paracrina/efectos de los fármacos , Radio (Anatomía)/efectos de los fármacos , Radio (Anatomía)/patología , Fusión Vertebral , Tetraciclina/farmacología , Cicatrización de Heridas/efectos de los fármacos
17.
Tissue Eng Part A ; 14(11): 1763-73, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18636943

RESUMEN

Stem cell-based bone tissue regeneration in the maxillofacial complex is a clinical necessity. Genetic engineering of mesenchymal stem cells (MSCs) to follow specific differentiation pathways may enhance the ability of these cells to regenerate and increase their clinical relevance. MSCs isolated from maxillofacial bone marrow (BM) are good candidates for tissue regeneration at sites of damage to the maxillofacial complex. In this study, we hypothesized that MSCs isolated from the maxillofacial complex can be engineered to overexpress the bone morphogenetic protein-2 gene and induce bone tissue regeneration in vivo. To demonstrate that the cells isolated from the maxillofacial complex were indeed MSCs, we performed a flow cytometry analysis, which revealed a high expression of mesenchyme-related markers and an absence of non-mesenchyme-related markers. In vitro, the MSCs were able to differentiate into osteogenic, chondrogenic, and adipogenic lineages. Gene delivery of the osteogenic gene BMP2 via an adenoviral vector revealed high expression levels of BMP2 protein that induced osteogenic differentiation of these cells in vitro and induced bone formation in an ectopic site in vivo. In addition, implantation of genetically engineered maxillofacial BM-derived MSCs into a mandibular defect led to regeneration of tissue at the site of the defect; this was confirmed by performing micro-computed tomography analysis. Histological analysis of the mandibles revealed osteogenic differentiation of implanted cells as well as bone tissue regeneration. We conclude that maxillofacial BM-derived MSCs can be genetically engineered to induce bone tissue regeneration in the maxillofacial complex and that this finding may be clinically relevant.


Asunto(s)
Regeneración Ósea/fisiología , Enfermedades Mandibulares/cirugía , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Huesos Faciales/citología , Femenino , Humanos , Lactante , Masculino , Enfermedades Mandibulares/fisiopatología , Maxilar/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Adulto Joven
18.
Tissue Eng Part A ; 14(10): 1709-20, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18620480

RESUMEN

Genetically modified mesenchymal stem cells (MSCs), overexpressing a BMP gene, have been previously shown to be potent inducers of bone regeneration. However, little was known of the chemical and intrinsic nanomechanical properties of this engineered bone. A previous study utilizing microcomputed tomography, back-scattered electron microscopy, energy-dispersive X-ray, nanoindentation, and atomic force microscopy showed that engineered ectopic bone, although similar in chemical composition and topography, demonstrated an elastic modulus range (14.6-22.1 GPa) that was less than that of the native bone (16.6-38.5 GPa). We hypothesized that these results were obtained due to the specific conditions that exist in an intramuscular ectopic implantation site. Here, we implanted MSCs overexpressing BMP-2 gene in an orthotopic site, a nonunion radial bone defect, in mice. The regenerated bone tissue was analyzed using the same methods previously utilized. The samples revealed high similarity between the engineered and native radii in chemical structure and elemental composition. In contrast to the previous study, nanoindentation data showed that, in general, the native bone exhibited a statistically similar elastic modulus values compared to that of the engineered bone, while the hardness was found to be marginally statistically different at 1000 muN and statistically similar at 7000 muN. We hypothesize that external loading, osteogenic cytokines and osteoprogenitors that exist in a fracture site could enhance the maturation of engineered bone derived from BMP-modified MSCs. Further studies should determine whether longer duration periods postimplantation would lead to increased bone adaptation.


Asunto(s)
Regeneración Ósea/fisiología , Huesos/fisiopatología , Células Madre Mesenquimatosas/citología , Animales , Fenómenos Biomecánicos , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Huesos/cirugía , Línea Celular , Femenino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/ultraestructura , Ratones , Microscopía de Fuerza Atómica , Microscopía Electrónica de Rastreo , Espectrometría Raman , Ingeniería de Tejidos/métodos , Tomógrafos Computarizados por Rayos X
19.
J Orthop Res ; 26(4): 522-30, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17985393

RESUMEN

Fluorescence molecular tomography (FMT) is a novel tomographic near-infrared (NIR) imaging modality that enables 3D quantitative determination of fluorochrome distribution in tissues of live small animals at any depth. This study demonstrates a noninvasive, quantitative method of monitoring engineered bone remodeling via FMT. Murine mesenchymal stem cells overexpressing the osteogenic gene BMP2 (mMSCs-BMP2) were implanted into the thigh muscle and into a radial nonunion bone defect model in C3H/HeN mice. Real-time imaging of bone formation was performed following systemic administration of the fluorescent bisphosphonate imaging agent OsteoSense, an hydroxyapatite-directed bone-imaging probe. The mice underwent imaging on days 7, 14, and 21 postimplantation. New bone formation at the implantation sites was quantified using micro-computed tomography (micro-CT) imaging. A higher fluorescent signal occurred at the site of the mMSC-BMP2 implants than that found in controls. Micro-CT imaging revealed a mass of mature bone formed in the implantation sites on day 21, a finding also confirmed by histology. These findings highlight the effectiveness of FMT as a functional platform for molecular imaging in the field of bone regeneration and tissue engineering.


Asunto(s)
Curación de Fractura/fisiología , Fracturas no Consolidadas/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Osteogénesis/fisiología , Fracturas del Radio/terapia , Animales , Femenino , Colorantes Fluorescentes , Fracturas no Consolidadas/diagnóstico por imagen , Fracturas no Consolidadas/patología , Ingeniería Genética , Ratones , Ratones Endogámicos C3H , Fracturas del Radio/diagnóstico por imagen , Fracturas del Radio/patología , Tomografía Computarizada por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA